Pancreatic ductal adenocarcinoma (PDA) is one of the deadliest cancers and is projected to soon be the second leading cause of cancer death. Median survival of PDA patients is 6–10 mo, with the majority of diagnoses occurring at later, metastatic stages that are refractory to treatment and accompanied by worsening prognoses. Glycosylation is one of the most common types of post-translational modifications. The complex landscape of glycosylation produces an extensive repertoire of glycan moieties, glycoproteins, and glycolipids, thus adding a dynamic and tunable level of intra- and intercellular signaling regulation. Aberrant glycosylation is a feature of cancer progression and influences a broad range of signaling pathways to promote disease onset and progression. However, despite being so common, the functional consequences of altered glycosylation and their potential as therapeutic targets remain poorly understood and vastly understudied in the context of PDA. In this review, the functionality of glycans as they contribute to hallmarks of PDA are highlighted as active regulators of disease onset, tumor progression, metastatic capability, therapeutic resistance, and remodeling of the tumor immune microenvironment. A deeper understanding of the functional consequences of altered glycosylation will facilitate future hypothesis-driven studies and identify novel therapeutic strategies in PDA.

Introduction

For the past 40 yr, survival rates for patients diagnosed with pancreatic ductal adenocarcinoma (PDA) have remained at <10% (Abbassi and Algül, 2019). Due to increasing incidence, difficulty in early diagnosis, and the refractory nature of advanced disease, PDA is projected to become the second leading cause of cancer death by 2040 (Abbassi and Algül, 2019). For years, gemcitabine has been the standard of care for PDA patients, providing median survival times of ∼6 mo (Abbassi and Algül, 2019). Although subsequent clinical trials demonstrated improved survival benefits of FOLFIRINOX (11.1 mo) and gemcitabine/nab-paclitaxel (8.5 mo), systemic chemotherapeutic regimens are rarely curative in PDA. Currently, surgical resection remains the only curative option, yet most patients are ineligible due to regional or distal metastases at the time of diagnosis (Kleeff et al., 2016).

In a concerted effort to provide more robust survival benefits for PDA patients, significant research strides in recent years have contributed substantially to our understanding of PDA. These studies have identified key driver mutations, signaling pathways, and tumor-microenvironment interactions that promote disease progression, regulate metastasis, and drive therapeutic resistance. Yet, the impact of glycosylation, one of the most common post-translational modifications which compounds the complexity of intra- and intercellular regulation, remains understudied in PDA. The paucity of such research in this area is driven by the vast aggregate complexity of glycans and their associated structures, the intricate mechanisms by which glycosylation and glycan assembly are regulated, and the lack of models that recapitulate glycosylation in human disease. For instance, only in recent years has a mouse model of pancreatic disease capable of producing the PDA-associated glycan CA19-9 been developed (Engle et al., 2019), providing evidence for the functional capacity of a glycan that was heretofore only considered a correlative biomarker of disease progression. Understanding the functional roles of glycosylation in PDA will be crucial for identifying both new diagnostic tools and therapeutic targets.

Landscape of glycosylation

Glycans exist as covalent linkages of saccharides in either free form or, more commonly, attached to proteins or lipids as glycoconjugates, thus forming glycoproteins, glycolipids, and proteoglycans (Ohtsubo and Marth, 2006; Fuster and Esko, 2005). In mammals, glycans are constructed from a combination of 10 monosaccharides (galactose, glucose, mannose, fucose, xylose, N-acetyl-galactosamine, N-acetyl-glucosamine, glucuronic acid, iduronic acid, and sialic acid), which are attached via α- or β-glycosidic bonds to form linear or branched structures (Kudelka et al., 2015). Glycan chain assembly is catalyzed by glycosyltransferases and glycosidases, which are encoded by 1–2% of the human genome (Taniguchi and Kizuka, 2015). Biochemically, glycans can be classified into families. N-glycans are linked via an amide bond to asparagine in the consensus Asn-X-Ser/Thr sequon (where X is any amino acid except proline). Structurally, N-glycans present as branched oligomannose, complex, or hybrid moieties. O-glycans are most commonly linked to the oxygen atom on serine or threonine, and in some cases tyrosine. These can be further subdivided into O-linked GlcNAc, as well as secreted or membrane glycoproteins decorated with O-glycans, the most common of which is the mucin-type (or GalNAc type) O-glycan. In contrast to N-glycans, no glycosite amino acid sequon has been identified for O-GalNAc-linked glycans (Kudelka et al., 2015).

Other glycan families include free or proteoglycan-conjugated long linear repeats of disaccharide units called glycosaminoglycans, ceramide-linked glycans called glycosphingolipids, and glycosylphosphatidylinositol-linked proteins (Fuster and Esko, 2005). Individual saccharides in glycan chains can be further modified, e.g., by phosphorylation, sulfation, and acetylation, increasing their structural diversity. Glycans are often found as secreted glycoconjugates or on the cell surface, forming a multifunctional layer of glycans termed the glycocalyx. The glycocalyx is responsible for a variety of biological functions including maintaining vascular permeability (Uchimido et al., 2019), immune cell recognition (Möckl, 2020), and crosstalk with the extracellular matrix (ECM; Kang et al., 2018).

In contrast to proteins, the sequence of glycan chains is not genetically encoded. Instead, glycan assembly and the formation of glycoconjugates are regulated by numerous factors, including the availability of nucleotide sugars that act as donor substrates (e.g., UDP-galactose, UDP-N-acetylglucosamine, GDP-fucose, and CMP-N-acetylneuraminic acid), acceptor substrates, and cofactors, as well as the glycosyltransferases and glycosidases necessary to catalyze such reactions. Further, each of these elements must be localized appropriately within the cell and secretory apparatus. This process is governed by molecular chaperones, endogenous lectins, and nucleotide sugar transporters (Taniguchi and Kizuka, 2015). The collection of enzymes that comprise the glycosylation machinery is primarily localized to the Golgi, with some also present in the endoplasmic reticulum (Taniguchi and Kizuka, 2015).

The vast diversity of the glycome, and the predominant presence of glycans on membranes and secreted proteins, positions glycoconjugates as mediators of multiple biological processes such as inter- and intracellular signaling, cell adhesion and motility, and immune regulation. Investigation of the functional roles of aberrant glycans has been difficult not only in part due to their complexity and families of glycosyltransferases with redundant functions, but also because of differences in the glycome between mouse models and humans. For example, mice endogenously display glycan epitopes not normally observed in humans, including α-Gal and Neu5Gc epitopes (Saleh et al., 2020; Altman and Gagneux, 2019). Conversely, sLea (CA19-9) is frequently upregulated in human pancreatic disease, but its synthesis is precluded in mice due to Fut3, the only enzyme capable of transferring the core α-1,4-fucose required for sLea synthesis, being a pseudogene in mice (Engle et al., 2019). Understanding glycosylation alterations, the regulatory aberrations underpinning them, and their consequences in the progression of malignant disease provide a promising and critical avenue to identify potential therapeutic vulnerabilities and deepen our understanding of cancer.

Aberrant glycosylation contributes to pancreatic cancer phenotypes

Evidence of altered glycosylation in cancer emerged in the 1970s, when glycopeptides isolated from transformed cell lines were observed to be larger than those from non-transformed cells (Buck et al., 1971). Around the same time, it was realized that many of the oncofetal and tumor-associated antigens used to detect and track disease progression were in fact carbohydrates (Feizi, 1985). Since then, substantial profiling of glycosylation alterations in various cancers has reshaped our understanding of how extensively glycosylation changes during the onset and progression of malignant disease (Munkley and Elliott, 2016; Gupta et al., 2020). Some of the most prevalent glycosylation irregularities described in PDA include dysregulated O-GlcNAc, increased sialylation and fucosylation, aberrantly branched O-glycan structures, and altered mucins (Qorri et al., 2020). In turn, these alterations impact various pro-tumorigenic signaling pathways, promote metastatic phenotypes, and remodel the tumor immune microenvironment. In the following sections, we describe the contributions of altered glycosylation in PDA to these phenotypes and highlight areas in which therapeutic vulnerabilities may exist.

Sustaining proliferative and pro-tumorigenic signaling

Intracellular signaling events regulated by glycosylation

Glycosylation-mediated intracellular signaling is primarily mediated by the O-linked β-N-acetylglucosamine (O-GlcNAc) modification. Levels of O-GlcNAc are increased in breast, colon, and pancreatic cancer (Nagel and Ball, 2015). O-GlcNAc modification alters the localization and activity of metabolic enzymes, histones, and transcriptional regulators (Fig. 1; Nagel and Ball, 2015). O-GlcNAc modification of transcription factors, including Sp1, β-catenin, SOX2, FOXO3, and YAP, regulates their nuclear translocation and activity, subsequently promoting gene expression programs that confer proliferative and anti-apoptotic cancer cell phenotypes (Banerjee et al., 2013; Jia et al., 2020; Sharma et al., 2019; Shin et al., 2018; Peng et al., 2017). Levels of O-GlcNAc are governed by the balance between O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), which add and remove UDP-GlcNAc to and from acceptor substrates, respectively. In addition to OGT and OGA, O-GlcNAcylation is sensitive to the nutritional and metabolic status of cells. The dense fibrotic and hypovascular nature of PDA leads to a hypoxic niche, increasing flux through glycolysis and glutamine metabolism (Guillaumond et al., 2014), pathways that converge toward the hexosamine biosynthetic pathway (HBP). The HBP is a branch of glycolysis responsible for the production of UDP-GlcNAc, a key substrate for glycosylation. HBP activity is increased in PDA and associated with poor survival (Jia et al., 2020). O-GlcNAcylation, hypoxia, and HPB converge in PDA. Hypoxia increases levels of OGT, OGA, and O-GlcNAc, and increases expression of glutamine–fructose-6-phosphate transaminase 1 (GFPT1), which catalyzes the first and the rate-limiting step of the HBP (Guillaumond et al., 2013). Pharmacologic targeting of the HBP using antagonists of GFPT disrupts O-GlcNAc protein modification, decreases cell proliferation, survival, and invasion, and increased starvation-induced apoptosis (Guillaumond et al., 2013; Jia et al., 2020). Thus, the centrality of the HBP in fueling aberrant protein glycosylation in cancer presents a promising therapeutic vulnerability.

In addition to O-GlcNAc, other types of glycosylation also impact intracellular signaling. Increased O-GalNAcylation, catalyzed by GalNAc transferases that are upregulated in PDA (Taniuchi et al., 2011; Sutherlin et al., 1997), renders malignant cells resistant to apoptosis by regulating the processing of caspase-3, -8, -9, and Bid. In addition to O-glycosylation, modification and stabilization of other intracellular proteins with sialic acid have been described, such as sialylation of HIF-1α (Jones et al., 2018).

Collectively, these studies highlight a dynamic interplay between the remodeled PDA tumor microenvironment and metabolically and genetically reprogrammed tumor cells that influence glycosylation-mediated intracellular signaling events and promote proliferative and anti-apoptotic phenotypes. By targeting the metabolic dependencies controlling these processes, pro-tumorigenic intracellular signaling mediated by aberrant O-GlcNAcylation can be abrogated.

Glycosylation mediated pro-tumorigenic cell surface receptor signaling

Like intracellular proteins, cell membrane receptor activity is also tunable via glycosylation modifications. Levels of the enzyme ST6Gal-I, which gives rise to α-2,6-sialylation, are increased in acinar-to-ductal metaplasia, pancreatitis, preinvasive pancreatic intraepithelial neoplasms, and PDA (Schultz et al., 2016). Increased α-2,6-sialylation of TNFR1 inhibits internalization and stabilizes signaling through AKT and NF-κB, conferring resistance to gemcitabine and TNF-induced apoptosis (Britain et al., 2017; Holdbrooks et al., 2018; Schultz et al., 2016; Chakraborty et al., 2018). Dimerization of several receptor tyrosine kinases and toll-like receptors are regulated by the sialidase neuraminidase 1 (Neu1), which cleaves terminal sialic acid residues from glycoproteins (Abdulkhalek and Szewczuk, 2013; Jayanth et al., 2010; Fig. 1). Specifically, ligand binding induces a receptor conformational change that results in MMP9 and Neu1 activation. This leads to the hydrolysis of terminal α-2,3-sialyl residues present on receptors, thereby removing steric hindrance and allowing RTK dimerization (Gilmour et al., 2013; Mathew et al., 2015; Mathew et al., 2016). Previous reports in lung cancer cells demonstrated that increased N-sialylation or N-fucosylation of EGFR suppresses dimerization and attenuates the activation of downstream signaling (Liu et al., 2011). Notably, EGFR activation is required for KRAS-induced pancreatic tumorigenesis (Ardito et al., 2012), underscoring a critical role for glycosylation in regulating epithelial transformation. Neu1-dependent RTK activation represents a targetable vulnerability, with aspirin and neuraminidase inhibitors such as Tamiflu capable of inhibiting Neu1 activity and preventing EGFR phosphorylation and downstream signaling (Qorri et al., 2020). Additionally, changes in N-glycosylation branching have been shown to increase metastatic dissemination of multiple cancer types and are correlated with PDA progression (Pan et al., 2014). In PDA, proteins with altered N-glycosylation sites or structures often play significant roles in pathways that increase metastasis such as the TGF-β and NF-κB signaling pathways (Pan et al., 2012).

Stabilization of cell membrane receptors is also crucial to their activity, and it is influenced by high molecular weight glycoproteins called mucins, which function canonically in lubrication, cell signaling, and barrier formation. Mucins have been reviewed extensively elsewhere (Suh et al., 2017). In PDA, the global expression of mucins is altered (Yamada et al., 2006). Mucins potentiate pro-tumorigenic signaling via a variety of mechanisms (Fig. 1). For instance, MUC16 interacts with EGF receptors to activate AKT and GSK3β signaling, an effect that can be reversed using anti-MUC16 antibodies (Thomas et al., 2021). Similarly, MUC4 interacts with and stabilizes HER2, and MUC4 knockdown in CD14/HPAF and Capan1 cells lead to reduced phosphorylation of the downstream effectors FAK and ERK (Chaturvedi et al., 2008). MUC20 interacts with MET, and knockdown of MUC20 in HPAC and HPAF-II cells decreased HGF-mediated phosphorylation of MET (Chen et al., 2018). Engagement of these mucin-mediated cell signaling programs results in pro-tumorigenic changes in cell behavior, including increases in cell viability, pancreatic stellate cell-induced migration and invasion, and in vivo orthotopic tumor growth (Chen et al., 2018). The resulting impact of mucins on patient outcomes is also clear. The expression of MUC4, MUC16, and MUC20 are all correlated with poor survival (Jonckheere and Van Seuningen, 2018; Ohya et al., 2017; Remmers et al., 2013; Haridas et al., 2011; Nagata et al., 2007; Ringel and Löhr, 2003; Yonezawa et al., 2002; Yonezawa and Sato, 1997). Notably, dense O-glycosylated mucins contribute to glycocalyx formation and can hinder the uptake of chemotherapies such as 5-fluorouracil, an effect that can be reversed by global inhibition of O-glycosylation with benzyl-α-GalNAc (Kalra and Campbell, 2009; Kalra and Campbell, 2007).

In addition to modifying receptors, the glycan decoration of ligands serves as another mechanism by which cell surface signaling is altered in the pancreatic disease. sLea/CA19-9 directly impacts the activation of EGFR and potentiates downstream signaling in pancreatic disease by decorating the EGFR ligand Fibulin-3 (Engle et al., 2019; Fig. 1). Mice genetically engineered to produce CA19-9 develop severe pancreatitis. In the KRAS-mutant context, CA19-9 elevation dramatically decreased survival (Engle et al., 2019). Importantly, pancreatitis severity and EGFR hyperactivation were attenuated by the treatment of mice with anti–CA19-9 blocking antibodies, including the fully human 5B1 clone (Weitzenfeld et al., 2019), highlighting a glycosylation-based therapeutic vulnerability in pancreatitis and PDA. 5B1 is currently being evaluated as an imaging modality and a therapeutic agent in clinical trials (Houghton et al., 2017; Lohrmann et al., 2019).

Role of glycosylation in PDA invasion and metastasis

Cell surface sialylation and fucosylation-mediated adhesion and invasion

Early investigations into how glycosylation changes influence metastasis focused on the sialylated and fucosylated Lewis blood group antigens that include sialyl-Lewis x (sLex) and sialyl-Lewis a (sLea, CA19-9). Upregulation of these glycan moieties in tissue and serum of PDA patients was discovered in the 1980s (Haglund et al., 1986; Kalthoff et al., 1986; Pour et al., 1988; Singh et al., 2015; Tang et al., 2015; Balmaña et al., 2015). Several subsequent ground-breaking publications demonstrated the potential of these antigens to increase the binding of circulating tumor cells to E-selectin on endothelial cells, potentially facilitating metastasis (Fig. 2; Phillips et al., 1990; Iwai et al., 1993; Kaji et al., 1995; Takada et al., 1991; Takada et al., 1993; Takada et al., 1995; Sawada et al., 1994). Conversely, engineering cells to divert glycosylation away from sLea production reduced E-selectin adhesion and metastases in transplanted mice (Aubert et al., 2000). Following metastatic progression, cell surface sialylation is also increased, with metastatic nodules displaying higher sialylated-GalNAc (sTn, a common antigen in cancer and the subject of several cancer vaccine studies) levels than the primary tumor or normal tissue (Itzkowitz et al., 1991; Fig. 2). Glycoengineering to increase metabolic flux through the sialic acid production pathway increases cell surface sialic acid concentration and consequently increases cellular binding to E- and L-selectin (Almaraz et al., 2012; Mathew et al., 2016), a key step in circulating tumor cell extravasation into distant tissues. Conversely, neuraminidase treatment to remove sialic acid on human PDA cells causes decreased adhesion to ECM (Sawada et al., 1993). Additionally, increases in tumor cell surface sialylation led to increases in the adhesive and invasive properties of tumor cells. Elevated expression of fucosyltransferases and sialyltransferases, such as FUT3, -5, -6, ST6Gal1, and ST3Gal3/4, similarly led to increases in cell motility and metastatic potential in both cell lines and mouse models (Gao et al., 2019; Bassagañas et al., 2014a; Bassagañas et al., 2014b; Pérez-Garay et al., 2013; Pérez-Garay et al., 2010; Hsieh et al., 2017; Britain et al., 2021).

In addition to sialyl Lewis antigens and changes in cell surface sialylation and fucosylation, alterations of other glycoproteins and glycan moieties, such as mucins and truncated O-glycan chains (Tn, sTn), potentiate PDA metastasis (Fig. 2; Gendler, 2001; Kohlgraf et al., 2003; Satoh et al., 2000; Hanson et al., 2016; Swanson et al., 2007; Mcdermott et al., 2001; Singh et al., 2007). In PDA, MUC1 interacts with both E- and P-selectins (Mcdermott et al., 2001) to facilitate extravasation and transmits signals into the cell from the surrounding microenvironment to induce increased invasion through stabilization and phosphorylation of FRA-1 and c-JUN (Hanson et al., 2016; Besmer et al., 2011). Similar results have also been reported for MUC16 in human cell lines (Muniyan et al., 2016). Meanwhile, the elevation of truncated O-glycan chains such as Tn and sTn occurs because of aberrations in O-glycosylation extension enzymes such as core synthase 3 and C1GALT1, resulting in enhanced cellular invasion, migration, and metastasis (Radhakrishnan et al., 2014; Hofmann et al., 2015; Chugh et al., 2018; Kuo et al., 2021b).

Collectively, these studies support a causal role for Lewis antigens, sialylation, fucosylation, mucins, and Tn/sTn in enhancing metastatic capability via interactions with ECM and glycan-binding lectins on the endothelium. However, the nuanced impacts of different sialyltransferases on PDA behavior underscore the complexity of glycosylation events and the need for careful consideration of how changes in glycosyltransferases affect the global glycome and cell function (Holst et al., 2017). Characterization of discrete glycosyltransferase and glycoconjugate contributions to different stages of the metastatic cascade will prove key to deepening our understanding of how to combat malignant spread.

Altered glycosylation in the extracellular space influences cell adhesion and metastasis

Glycoconjugates often impact cellular motility, adhesion, and invasive capacity through their action in the extracellular space. For example, glycosaminoglycans (GAGs) are long polysaccharide chains of repeating two-sugar subunits that can have profound impacts on tumor interstitial pressure. The high anionic charge of GAGs alters the water content, and thus pressure, of tissues and tumors (Vedadghavami et al., 2020), which has implications for drug delivery (Provenzano and Hingorani, 2013). Heparan sulfate GAGs (HSGAGs), such as Syndecans (SDC) and glypicans, are bound to a proteoglycan core attached to the cell membrane and are involved in tumor development, cell adhesion, and metastasis (Nagarajan et al., 2018). SDC are often upregulated in gastric and colon cancers, but Syndecan-1 (SDC1) is upregulated only in pancreatic cancer, where it correlates with invasion of cancer and a poor prognosis (Conejo et al., 2000; Yao et al., 2019). Increased levels of heparanase (HPA) in pancreatic cancer increase metastatic potential by facilitating shedding of SDC1 as well as activation of the PI3K/AKT pathway and the epithelial-to-mesenchymal transition (Yang et al., 2007; Chen et al., 2020). Both SDC1 and SDC2 cooperate with KRAS mutation to promote PDA metastasis through activation of SRC kinase and phosphorylation of ERK (Yao et al., 2019; De Oliveira et al., 2012). Membrane-anchored glypicans modulate chemokine and growth factor signaling (Kleeff et al., 1998). Glypican-1 increases PDA invasion and metastasis through the modulation of FGF2 signaling (Aikawa et al., 2008). Glypican-4 has also been implicated in PDA metastasis and recurrence by increasing stemness markers through the WNT/β-catenin pathway within tumors (Cao et al., 2018). The basement membrane HSGAG Perlecan has also been shown to be upregulated in pancreatic cancer and has a direct effect on cancer-associated fibroblast populations. The depletion of Perlecan abrogated the CAF-induced pro-metastatic niche in mice, resulting in increased survival (Vennin et al., 2019).

Unlike membrane-tethered HSGAGs, hyaluronan (hyaluronic acid or HA) is a secreted GAG that is not bound to the cell surface. The effects of HA on migration, invasion, and metastasis have been extensively reviewed elsewhere (Sato et al., 2016). HA binds to its cell surface membrane receptor CD44, which has many isoforms and splice variants, some of which are only expressed in cancer (Sato et al., 2016). HA binding of CD44 results in increased PI3K signaling, invasion, and metastasis (Teranishi et al., 2009). In PDA, HA contributes to increased interstitial fluid pressure, which collapses vasculature and creates a barrier to treatment (Provenzano et al., 2012). Enzymatic ablation of HA decreases tumor and metastatic burden in KPC mice (Jacobetz et al., 2013). In light of the significant abundance and potential contribution to pancreatic cancer of HA, a clinical trial was initiated to determine whether the disruption of HA could improve patient outcomes. However, therapeutic targeting of HA in combination with nab-paclitaxel/gemcitabine was insufficient to improve patient outcomes in a Phase II clinical trial (Hingorani et al., 2018). The complex interplay between altered glycoconjugates and the tumor microenvironment will likely necessitate combinatorial approaches to target key prosurvival networks in PDA.

The compendium of both free and membrane-bound proteoglycans, glycosaminoglycans, glycolipids, and glycosylated proteins comprises the glycocalyx, which presents as a mesh that covers the cell and is at the interface of cell–cell and cell–matrix signaling. The glycocalyx plays key functional roles in cell morphology, regulation of membrane protein diffusion, and immune cell regulation (Möckl, 2020). Alterations in the glycocalyx are capable of contributing to tumor progression by modulating cell membrane protein turnover, membrane topology, and cell morphology to facilitate biophysical interactions with the surrounding microenvironment (Al-Aghbar et al., 2022; Cruz-Chu et al., 2014; Möckl, 2020). Changes in the glycocalyx are known to be important in various types of cancers, yet there is still more to learn regarding its role in pancreatic cancer. Recently, doxycycline-inducible KRAS mutant cell lines were utilized to visualize the glycocalyx of PDA cells, demonstrating a mutant KRAS-dependent increase in glycocalyx size, potentially decreasing the adhesion of cells to the surrounding stroma or matrix (Möckl, 2020). The glycocalyx also has physical effects on integrins in normal and tumor contexts by changing pH or physical restriction of integrins to a specific region. While this phenomenon requires further study in pancreatic cancer, it is known that integrins are aberrantly glycosylated in cancer and are critical signaling integration hubs through which cell interactions with the matrix are transmitted. Several studies have shown that in PDA, altered glycosylation of integrins changes their adhesion properties. These integrins are no longer able to bind as effectively to the matrix or E-cadherin, thereby increasing the invasive potential of tumor cells (Kuo et al., 2021a; Bassaganas et al., 2014).

Glycoconjugates in the PDA immune microenvironment

Aberrant glycosylation facilitates tumor immune escape

Glycans in cancer have long been correlated with suppressed anti-tumor immune responses (Varki et al., 2015). Glycans and glycoproteins often lie at the cell–cell interface and frequently comprise pathogen- and damage-associated molecular patterns which activate the innate immune system. By altering the cell surface glycan “code,” transformed cells can evade immune detection. Indeed, increased expression of ST3Gal1 and -4 leads to increased sialylation on PDA cells that induces monocyte differentiation into immunosuppressive tumor-associated macrophages via myeloid receptors Siglec-7 and -9 (Rodriguez et al., 2021; Fig. 3). Reciprocally, inflammatory cytokines present in the microenvironmental milieu, such as IL1β, IL6, and TNFα, upregulate PDA cell expression of ST3GAL3-4, FUT1-2, and FUT6, resulting in increased sLex, SLey, and α2,6-sialic acid levels, suggesting that glycosylation of PDA cells is modulated by inflammatory microenvironments (Bassagañas et al., 2015; Fig. 3).

MUC1 is overexpressed in colon, breast, ovarian, lung, and pancreatic cancers (Gendler, 2001; Sharma and Allison, 2015; Hanson and Hollingsworth, 2016; Apostolopoulos and McKenzie, 2017). Expression of MUC1 results in elevated expression of multiple proinflammatory cytokines and concomitant increases in regulatory T cell and myeloid suppressor cell populations in pancreatic tumors (Tinder et al., 2008; Fig. 3). Furthermore, cancer cells that express MUC1 display increased sialyl-Tn antigen (Dalziel et al., 2001; Gill et al., 2011; Reis et al., 1998), and alterations in MUC1 affect immune detection and increase macrophage activation through binding to sialylated MUC1, promoting tumor growth (Saeland et al., 2007; Nath et al., 1999; Cascio and Finn, 2016). Exposed cryptic peptide epitopes, as a result of truncated glycosylation, can be recognized by antibodies and are ideal targets for therapy. The potential utility of various MUC1-based vaccine strategies has shown promise in eliciting immune responses in preclinical trials, leading to clinical trials in several cancers, including PDA, that have demonstrated a range of effects on humoral and T cell responses and patient survival (Gao et al., 2020).

Aberrant glycosylation and splicing of mucins can result in longer glycan chains, physically hindering immune interaction with cancer cells and supporting tumor progression and immunosuppression by shielding cancer-associated epitopes (Fig. 3; Jahan et al., 2018). MUC4 provides steric hindrance via its bulky, glycosylated extracellular region to mask immune cell surface antigens and shield tumor cells from immune recognition, thus enhancing survival and extravasation of metastasized tumor cells via interaction with macrophages and hematopoietic progenitors (Rowson-Hodel et al., 2018). Truncated mucin-type O-glycans are observed in many epithelial cancer cells and premalignant lesions in adenocarcinomas (Itzkowitz et al., 1991; Ching et al., 1993; Lyubsky et al., 1988; Kim et al., 2002; Tarp and Clausen, 2008; Stanley, 2011). In an immunological context, the truncated glycans Tn and sTn prevent O-linked glycan elongation beyond the initial GalNAc residue, increasing sensitivity to NK cell–mediated antibody-dependent cellular cytotoxicity (ADCC) and cytotoxic T-lymphocyte–mediated killing (Madsen et al., 2013). Understanding how aberrant glycosylation contributes to the immunosuppressive microenvironment of PDA will be critical in the design of therapeutic strategies targeting these alterations.

Leveraging glyco-epitopes in cancer immunotherapy

Recent years have demonstrated effective reactivation of T cell immunity via blockade of the immune checkpoint PD-1/PD-L1 pathway, thereby improving survival in patients with various types of cancer (Chen and Han, 2015; Sharma and Allison, 2015; Topalian et al., 2016; Qin et al., 2019). Unfortunately, many patients only partially respond to PD-1/PD-L1 inhibition (Zou et al., 2016; Topalian et al., 2015). It is critical to understand how post-translational modifications, such as N-linked glycosylation, regulate PD-L1. PD-L1 is N-glycosylated in melanoma as well as breast, lung, and colon cancer (Li et al., 2016). N-linked glycosylation of PD-L1 enhances its protein stability and plays a vital role in PD-1/PD-L1–mediated tumor immunosuppressive function (Li et al., 2016; Hsu et al., 2018; Wang et al., 2018; Cha et al., 2019). Glycosylated PD-L1 suppresses T cell activity, whereas non-glycosylated PD-L1 exhibits less immunosuppressive activity, resulting in slower tumor growth (Li et al., 2018). It will be critical to extend these studies to PDA to understand if altered PD-L1 glycosylation contributes to immunosuppression and poor immune checkpoint blockade therapy response rates.

Current strides in immunotherapies and anti-tumoral immunity involve utilizing glycosite-targeting antibodies to block immunosuppressive interactions and glycoepitope-induced dendritic cell responses to boost T lymphocyte activation in PDA and other cancers. To determine how O-glycosylation impacts steric hindrance by mucins, the binding of an anti-MUC1 monoclonal antibody HMFG-2 was assessed in breast, colon, and pancreatic cancer cell lines. HMFG-2 reactivity against MUC1 increased following inhibition of O-glycosylation using benzyl-α-N-acetylgalactosamide to prevent glycan chain elongation. De-sialylation of cell surfaces also improved access to MUC1 (Ho et al., 1995). The immunodominant DTR (Aspartic Acid-Threonine-Arginine) motif of MUC1, which comprises part of the exposed core peptide resulting from aberrantly truncated glycan chains on MUC1, has been the primary target of many immunotherapeutic interventions, and prior work had uncovered enhanced antibody binding to peptides O-glycosylated with GalNAc in the DTR motif (Thr-10) compared to non-glycosylated peptides (Karsten et al., 1998; Karsten et al., 2004). Pankomab, an antibody targeting this carbohydrate-induced conformational tumor-associated epitope on MUC1, features the highest glycosylation dependency and strongest additive binding effect compared to past MUC1 antibodies (Danielczyk et al., 2006). Upon binding, it induces an elevated cytotoxic T-lymphocyte response on tumor cells, and MUC1–chimeric antigen receptor T cells show target-specific cytotoxicity and tumor inhibition in pancreatic cancer xenograft models (Cai et al., 2015).

Immunotherapeutic strategies that target other glycoproteins are also showing promise. CD24, a cell surface sialoglycoprotein expressed by most B lymphocytes, is overexpressed in many human carcinomas. Prior studies have shown that CD24-specific monoclonal SWA11 therapy effectively impeded lung and pancreatic tumor growth in xenotransplanted mice (Bretz et al., 2012). Furthermore, SWA11 therapy not only decreased lung and ovarian carcinoma tumor cell proliferation and affected the tumor microenvironment cytokine milieu in mice, but also results in increased tumor infiltration of macrophages (Salnikov et al., 2013). The cumulative tumor-promoting and restraining effects of CD24 targeting by monoclonal therapy will be critical to explore in autochthonous mouse models of cancer in the context of standard chemotherapeutic intervention.

Other novel approaches to cancer immunotherapy involve postsurgical vaccination with tumor antigen-loaded dendritic cells (Palucka and Banchereau, 2013; Dodson et al., 2011). Fucose-rich glycovariants of bile salt–dependent lipase (BSDL) are expressed during pancreatic tumorigenesis (pBSDL-J28; Mas et al., 1997). Treating mice with dendritic cells loaded with pBSDL-J28 induced T-lymphocyte activation, prevented Panc02 tumor development, and provided long-term protection against Panc02 tumor formation (Collignon et al., 2015). Overall, glycosylation alterations have enormous potential in immunotherapy, but further exploration is necessary to understand how to leverage these targets for the treatment of PDA.

Conclusions looking forward

Immense strides in glycobiology research have expanded our understanding of glycosylation in cancer biology and have cemented aberrant glycosylation as a key feature in tumor onset and progression. Associations between the presence of aberrant glycan moieties and malignant disease progression are well-documented, with some demonstrating clinical relevance serving as biomarkers of disease progression. Given the dynamic and complex mosaic of glycans and glycoconjugates and their multifaceted regulation, continuing research is required to further understand their functional and causative roles during discrete stages of disease progression. This understanding will reveal vulnerabilities in proliferative signaling, metastatic capabilities, and immune escape that can serve as promising combinatorial therapeutic targets. The emergence of model systems that faithfully recapitulate the human glycome and the heterogeneity observed across patients, such as mouse models of pancreatic disease that produce CA19-9, tissue slices, and human patient-derived organoids, will be crucial in this effort and may reveal critical avenues for therapeutic intervention.

We would like to thank Amy Cao for the figure illustrations.

The authors are supported by the National Institutes of Health, National Cancer Institute grants R00CA024725 (D.D. Engle), P30DK120515 (D.D. Engle), T32CA009370 (J.C. Lumibao), and T32GM133351 (J. Hsu), Salkexcellerator (J.R. Tremblay), the Pancreatic Cancer Action Network 19-20-ENGL (D.D. Engle), the American Association for Cancer Research and the Lustgarten Foundation 21-20-67-ENGL (D.D. Engle), the University of California Tobacco-Related Disease Research Program T31KT1898 (D.D. Engle), and Padres Pedal the Cause/C3 #PTC2020 (D.D. Engle). We are grateful to the Rose Hills Foundation, Mission Cure Capital, LLC, the Mark Foundation for Cancer Research, and the Emerald Foundation, Inc. for supporting D.D. Engle.

Author contributions: J.C. Lumibao, J.R. Tremblay, J. Hsu, and D.D. Engle wrote, reviewed, and edited the manuscript.

Abbassi
,
R.
, and
H.
Algül
.
2019
.
Palliative chemotherapy in pancreatic cancer-treatment sequences
.
Transl. Gastroenterol. Hepatol.
4
:
56
.
Abdulkhalek
,
S.
, and
M.R.
Szewczuk
.
2013
.
Neu1 sialidase and matrix metalloproteinase-9 cross-talk regulates nucleic acid-induced endosomal TOLL-like receptor-7 and -9 activation, cellular signaling and pro-inflammatory responses
.
Cell Signal
.
25
:
2093
2105
.
Aikawa
,
T.
,
C.A.
Whipple
,
M.E.
Lopez
,
J.
Gunn
,
A.
Young
,
A.D.
Lander
, and
M.
Korc
.
2008
.
Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells
.
J. Clin. Invest.
118
:
89
99
.
Al-Aghbar
,
M.A.
,
A.K.
Jainarayanan
,
M.L.
Dustin
, and
S.R.
Roffler
.
2022
.
The interplay between membrane topology and mechanical forces in regulating T cell receptor activity
.
Commun. Biol.
5
:
40
.
Almaraz
,
R.T.
,
Y.
Tian
,
R.
Bhattarcharya
,
E.
Tan
,
S.H.
Chen
,
M.R.
Dallas
,
L.
Chen
,
Z.
Zhang
,
H.
Zhang
,
K.
Konstantopoulos
, and
K.J.
Yarema
.
2012
.
Metabolic flux increases glycoprotein sialylation: Implications for cell adhesion and cancer metastasis
.
Mol. Cell Proteomics
.
11
:
M112.017558
.
Altman
,
M.O.
, and
P.
Gagneux
.
2019
.
Absence of Neu5Gc and presence of anti-Neu5Gc antibodies in humans-an evolutionary perspective
.
Front. Immunol.
10
:
789
.
Apostolopoulos
,
V.
, and
I.F.C.
Mckenzie
.
2017
.
Cellular mucins: Targets for immunotherapy
.
Crit. Rev. Immunol.
37
:
421
437
.
Ardito
,
C.M.
,
B.M.
Grüner
,
K.K.
Takeuchi
,
C.
Lubeseder-Martellato
,
N.
Teichmann
,
P.K.
Mazur
,
K.E.
Delgiorno
,
E.S.
Carpenter
,
C.J.
Halbrook
,
J.C.
Hall
, et al
.
2012
.
EGF receptor is required for KRAS-induced pancreatic tumorigenesis
.
Cancer Cell
.
22
:
304
317
.
Aubert
,
M.
,
L.
Panicot
,
C.
Crotte
,
P.
Gibier
,
D.
Lombardo
,
M.O.
Sadoulet
, and
E.
Mas
.
2000
.
Restoration of alpha(1,2) fucosyltransferase activity decreases adhesive and metastatic properties of human pancreatic cancer cells
.
Cancer Res.
60
:
1449
1456
Balmaña
,
M.
,
A.
Sarrats
,
E.
Llop
,
S.
Barrabés
,
R.
Saldova
,
M.J.
Ferri
,
J.
Figueras
,
E.
Fort
,
R.
De Llorens
,
P.M.
Rudd
, and
R.
Peracaula
.
2015
.
Identification of potential pancreatic cancer serum markers: increased sialyl-Lewis X on ceruloplasmin
.
Clin. Chim. Acta.
442
:
56
62
.
Banerjee
,
S.
,
V.
Sangwan
,
O.
Mcginn
,
R.
Chugh
,
V.
Dudeja
,
S.M.
Vickers
, and
A.K.
Saluja
.
2013
.
Triptolide-induced cell death in pancreatic cancer is mediated by O-GlcNAc modification of transcription factor Sp1
.
J. Biol. Chem.
288
:
33927
33938
.
Bassagañas
,
S.
,
H.
Allende
,
L.
Cobler
,
M.R.
Ortiz
,
E.
Llop
,
C.
De Bolós
, and
R.
Peracaula
.
2015
.
Inflammatory cytokines regulate the expression of glycosyltransferases involved in the biosynthesis of tumor-associated sialylated glycans in pancreatic cancer cell lines
.
Cytokine
.
75
:
197
206
.
Bassaganas
,
S.
,
S.
Carvalho
,
A.M.
Dias
,
M.
Perez-Garay
,
M.R.
Ortiz
,
J.
Figueras
,
C.A.
Reis
,
S.S.
Pinho
, and
R.
Peracaula
.
2014
.
Pancreatic cancer cell glycosylation regulates cell adhesion and invasion through the modulation of α2β1 integrin and E-cadherin function
.
PLoS One
.
9
:e98595.
Bassagañas
,
S.
,
S.
Carvalho
,
A.M.
Dias
,
M.
Pérez-Garay
,
M.R.
Ortiz
,
J.
Figueras
,
C.A.
Reis
,
S.S.
Pinho
, and
R.
Peracaula
.
2014a
.
Pancreatic cancer cell glycosylation regulates cell adhesion and invasion through the modulation of α2β1 integrin and E-cadherin function
.
PLoS One
.
9
:e98595.
Bassagañas
,
S.
,
M.
Pérez-Garay
, and
R.
Peracaula
.
2014b
.
Cell surface sialic acid modulates extracellular matrix adhesion and migration in pancreatic adenocarcinoma cells
.
Pancreas
.
43
:
109
117
.
Besmer
,
D.M.
,
J.M.
Curry
,
L.D.
Roy
,
T.L.
Tinder
,
M.
Sahraei
,
J.
Schettini
,
S.I.
Hwang
,
Y.Y.
Lee
,
S.J.
Gendler
, and
P.
Mukherjee
.
2011
.
Pancreatic ductal adenocarcinoma mice lacking mucin 1 have a profound defect in tumor growth and metastasis
.
Cancer Res.
71
:
4432
4442
.
Bretz
,
N.
,
A.
Noske
,
S.
Keller
,
N.
Erbe-Hofmann
,
T.
Schlange
,
A.V.
Salnikov
,
G.
Moldenhauer
,
G.
Kristiansen
, and
P.
Altevogt
.
2012
.
CD24 promotes tumor cell invasion by suppressing tissue factor pathway inhibitor-2 (TFPI-2) in a c-Src-dependent fashion
.
Clin. Exp. Metastasis
.
29
:
27
38
.
Britain
,
C.M.
,
N.
Bhalerao
,
A.D.
Silva
,
A.
Chakraborty
,
D.J.
Buchsbaum
,
M.R.
Crowley
,
D.K.
Crossman
,
Y.J.K.
Edwards
, and
S.L.
Bellis
.
2021
.
Glycosyltransferase ST6Gal-I promotes the epithelial to mesenchymal transition in pancreatic cancer cells
.
J. Biol. Chem.
296
:
100034
.
Britain
,
C.M.
,
K.A.
Dorsett
, and
S.L.
Bellis
.
2017
.
The glycosyltransferase ST6Gal-I protects tumor cells against serum growth factor withdrawal by enhancing survival signaling and proliferative potential
.
J. Biol. Chem.
292
:
4663
4673
.
Buck
,
C.A.
,
M.C.
Glick
, and
L.
Warren
.
1971
.
Glycopeptides from the surface of control and virus-transformed cells
.
Science
.
172
:
169
171
.
Cai
,
H.
,
B.
Palitzsch
,
S.
Hartmann
,
N.
Stergiou
,
H.
Kunz
,
E.
Schmitt
, and
U.
Westerlind
.
2015
.
Antibody induction directed against the tumor-associated MUC4 glycoprotein
.
Chembiochem
.
16
:
959
967
.
Cao
,
J.
,
J.
Ma
,
L.
Sun
,
J.
Li
,
T.
Qin
,
C.
Zhou
,
L.
Cheng
,
K.
Chen
,
W.
Qian
,
W.
Duan
, et al
.
2018
.
Targeting glypican-4 overcomes 5-FU resistance and attenuates stem cell-like properties via suppression of Wnt/β-catenin pathway in pancreatic cancer cells
.
J. Cell Biochem.
119
:
9498
9512
.
Cascio
,
S.
, and
O.J.
Finn
.
2016
.
Intra- and extra-cellular events related to altered glycosylation of MUC1 promote chronic inflammation, tumor progression, invasion, and metastasis
.
Biomolecules
.
6
:
E39
.
Cha
,
J.H.
,
L.C.
Chan
,
C.W.
Li
,
J.L.
Hsu
, and
M.C.
Hung
.
2019
.
Mechanisms controlling PD-L1 expression in cancer
.
Mol. Cell
.
76
:
359
370
.
Chakraborty
,
A.
,
K.A.
Dorsett
,
H.Q.
Trummell
,
E.S.
Yang
,
P.G.
Oliver
,
J.A.
Bonner
,
D.J.
Buchsbaum
, and
S.L.
Bellis
.
2018
.
ST6Gal-I sialyltransferase promotes chemoresistance in pancreatic ductal adenocarcinoma by abrogating gemcitabine-mediated DNA damage
.
J. Biol. Chem.
293
:
984
994
.
Chaturvedi
,
P.
,
A.P.
Singh
,
S.
Chakraborty
,
S.C.
Chauhan
,
S.
Bafna
,
J.L.
Meza
,
P.K.
Singh
,
M.A.
Hollingsworth
,
P.P.
Mehta
, and
S.K.
Batra
.
2008
.
MUC4 mucin interacts with and stabilizes the HER2 oncoprotein in human pancreatic cancer cells
.
Cancer Res.
68
:
2065
2070
.
Chen
,
L.
, and
X.
Han
.
2015
.
Anti-PD-1/PD-L1 therapy of human cancer: Past, present, and future
.
J. Clin. Invest.
125
:
3384
3391
.
Chen
,
S.T.
,
T.C.
Kuo
,
Y.Y.
Liao
,
M.C.
Lin
,
Y.W.
Tien
, and
M.C.
Huang
.
2018
.
Silencing of MUC20 suppresses the malignant character of pancreatic ductal adenocarcinoma cells through inhibition of the HGF/MET pathway
.
Oncogene
.
37
:
6041
6053
.
Chen
,
X.
,
H.
Zhao
,
C.
Chen
,
J.
Li
,
J.
He
,
X.
Fu
, and
H.
Zhao
.
2020
.
The HPA/SDC1 axis promotes invasion and metastasis of pancreatic cancer cells by activating EMT via FGF2 upregulation
.
Oncol. Lett.
19
:
211
220
.
Ching
,
C.K.
,
S.W.
Holmes
,
G.K.
Holmes
, and
R.G.
Long
.
1993
.
Comparison of two sialosyl-Tn binding monoclonal antibodies (MLS102 and B72.3) in detecting pancreatic cancer
.
Gut
.
34
:
1722
1725
.
Chugh
,
S.
,
S.
Barkeer
,
S.
Rachagani
,
R.K.
Nimmakayala
,
N.
Perumal
,
R.
Pothuraju
,
P.
Atri
,
S.
Mahapatra
,
I.
Thapa
,
G.A.
Talmon
, et al
.
2018
.
Disruption of C1galt1 gene promotes development and metastasis of pancreatic adenocarcinomas in mice
.
Gastroenterology
.
155
:
1608
1624
.
Collignon
,
A.
,
A.T.
Perles-Barbacaru
,
S.
Robert
,
F.
Silvy
,
E.
Martinez
,
I.
Crenon
,
S.
Germain
,
S.
Garcia
,
A.
Viola
,
D.
Lombardo
, et al
.
2015
.
A pancreatic tumor-specific biomarker characterized in humans and mice as an immunogenic onco-glycoprotein is efficient in dendritic cell vaccination
.
Oncotarget
.
6
:
23462
23479
.
Conejo
,
J.R.
,
J.
Kleeff
,
A.
Koliopanos
,
K.
Matsuda
,
Z.W.
Zhu
,
H.
Goecke
,
N.
Bicheng
,
A.
Zimmermann
,
M.
Korc
,
H.
Friess
, and
M.W.
Buchler
.
2000
.
Syndecan-1 expression is up-regulated in pancreatic but not in other gastrointestinal cancers
.
Int. J. Cancer
.
88
:
12
20
.
Cruz-Chu
,
E.R.
,
A.
Malafeev
,
T.
Pajarskas
,
I.V.
Pivkin
, and
P.
Koumoutsakos
.
2014
.
Structure and response to flow of the glycocalyx layer
.
Biophys. J.
106
:
232
243
.
Dalziel
,
M.
,
C.
Whitehouse
,
I.
Mcfarlane
,
I.
Brockhausen
,
S.
Gschmeissner
,
T.
Schwientek
,
H.
Clausen
,
J.M.
Burchell
, and
J.
Taylor-Papadimitriou
.
2001
.
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.
276
:
11007
11015
.
Danielczyk
,
A.
,
R.
Stahn
,
D.
Faulstich
,
A.
Löffler
,
A.
Märten
,
U.
Karsten
, and
S.
Goletz
.
2006
.
PankoMab: A potent new generation anti-tumour MUC1 antibody
.
Cancer Immunol. Immunother.
55
:
1337
1347
.
De Oliveira
,
T.
,
I.
Abiatari
,
S.
Raulefs
,
D.
Sauliunaite
,
M.
Erkan
,
B.
Kong
,
H.
Friess
,
C.W.
Michalski
, and
J.
Kleeff
.
2012
.
Syndecan-2 promotes perineural invasion and cooperates with K-ras to induce an invasive pancreatic cancer cell phenotype
.
Mol. Cancer
.
11
:
19
.
Dodson
,
L.F.
,
W.G.
Hawkins
, and
P.
Goedegebuure
.
2011
.
Potential targets for pancreatic cancer immunotherapeutics
.
Immunotherapy
.
3
:
517
537
.
Engle
,
D.D.
,
H.
Tiriac
,
K.D.
Rivera
,
A.
Pommier
,
S.
Whalen
,
T.E.
Oni
,
B.
Alagesan
,
E.J.
Lee
,
M.A.
Yao
,
M.S.
Lucito
, et al
.
2019
.
The glycan CA19-9 promotes pancreatitis and pancreatic cancer in mice
.
Science
.
364
:
1156
1162
.
Feizi
,
T.
1985
.
Demonstration by monoclonal antibodies that carbohydrate structures of glycoproteins and glycolipids are onco-developmental antigens
.
Nature
.
314
:
53
57
.
Fuster
,
M.M.
, and
J.D.
Esko
.
2005
.
The sweet and sour of cancer: Glycans as novel therapeutic targets
.
Nat. Rev. Cancer
.
5
:
526
542
.
Gao
,
H.F.
,
Q.Y.
Wang
,
K.
Zhang
,
L.Y.
Chen
,
C.S.
Cheng
,
H.
Chen
,
Z.Q.
Meng
,
S.M.
Zhou
, and
Z.
Chen
.
2019
.
Overexpressed N-fucosylation on the cell surface driven by FUT3, 5, and 6 promotes cell motilities in metastatic pancreatic cancer cell lines
.
Biochem. Biophys. Res. Commun.
511
:
482
489
.
Gao
,
T.
,
Q.
Cen
, and
H.
Lei
.
2020
.
A review on development of MUC1-based cancer vaccine
.
Biomed. Pharmacother.
132
:
110888
.
Gendler
,
S.J.
2001
.
MUC1, the renaissance molecule
.
J. Mammary Gland Biol. Neoplasia
.
6
:
339
353
.
Gill
,
D.J.
,
H.
Clausen
, and
F.
Bard
.
2011
.
Location, location, location: New insights into O-GalNAc protein glycosylation
.
Trends Cell Biol.
21
:
149
158
.
Gilmour
,
A.M.
,
S.
Abdulkhalek
,
T.S.W.
Cheng
,
F.
Alghamdi
,
P.
Jayanth
,
L.K.
O’shea
,
O.
Geen
,
L.A.
Arvizu
, and
M.R.
Szewczuk
.
2013
.
A novel epidermal growth factor receptor-signaling platform and its targeted translation in pancreatic cancer
.
Cell Signal
.
25
:
2587
2603
.
Guillaumond
,
F.
,
J.L.
Iovanna
, and
S.
Vasseur
.
2014
.
Pancreatic tumor cell metabolism: Focus on glycolysis and its connected metabolic pathways
.
Arch. Biochem. Biophys.
545
:
69
73
.
Guillaumond
,
F.
,
J.
Leca
,
O.
Olivares
,
M.N.
Lavaut
,
N.
Vidal
,
P.
Berthezène
,
N.J.
Dusetti
,
C.
Loncle
,
E.
Calvo
,
O.
Turrini
, et al
.
2013
.
Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma
.
Proc. Natl. Acad. Sci. USA
.
110
:
3919
3924
.
Gupta
,
R.
,
F.
Leon
,
C.M.
Thompson
,
R.
Nimmakayala
,
S.
Karmakar
,
P.
Nallasamy
,
S.
Chugh
,
D.R.
Prajapati
,
S.
Rachagani
,
S.
Kumar
, and
M.P.
Ponnusamy
.
2020
.
Global analysis of human glycosyltransferases reveals novel targets for pancreatic cancer pathogenesis
.
Br. J. Cancer
.
122
:
1661
1672
.
Haglund
,
C.
,
J.
Lindgren
,
P.J.
Roberts
, and
S.
Nordling
.
1986
.
Gastrointestinal cancer-associated antigen CA 19-9 in histological specimens of pancreatic tumours and pancreatitis
.
Br. J. Cancer
.
53
:
189
195
.
Hanson
,
R.L.
,
R.B.
Brown
,
M.M.
Steele
,
P.M.
Grandgenett
,
J.A.
Grunkemeyer
, and
M.A.
Hollingsworth
.
2016
.
Identification of FRA-1 as a novel player in pancreatic cancer in cooperation with a MUC1: ERK signaling axis
.
Oncotarget
.
7
:
39996
40011
.
Hanson
,
R.L.
, and
M.A.
Hollingsworth
.
2016
.
Functional consequences of differential O-glycosylation of MUC1, MUC4, and MUC16 (downstream effects on signaling)
.
Biomolecules
.
6
:
E34
.
Haridas
,
D.
,
S.
Chakraborty
,
M.P.
Ponnusamy
,
I.
Lakshmanan
,
S.
Rachagani
,
E.
Cruz
,
S.
Kumar
,
S.
Das
,
S.M.
Lele
,
J.M.
Anderson
, et al
.
2011
.
Pathobiological implications of MUC16 expression in pancreatic cancer
.
PLoS One
.
6
:e26839.
Hingorani
,
S.R.
,
L.
Zheng
,
A.J.
Bullock
,
T.E.
Seery
,
W.P.
Harris
,
D.S.
Sigal
,
F.
Braiteh
,
P.S.
Ritch
,
M.M.
Zalupski
,
N.
Bahary
, et al
.
2018
.
HALO 202: Randomized Phase II study of PEGPH20 plus Nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma
.
J. Clin. Oncol.
36
:
359
366
.
Ho
,
J.J.
,
S.
Cheng
, and
Y.S.
Kim
.
1995
.
Access to peptide regions of a surface mucin (MUC1) is reduced by sialic acids
.
Biochem. Biophys. Res. Commun.
210
:
866
873
.
Hofmann
,
B.T.
,
L.
Schlüter
,
P.
Lange
,
B.
Mercanoglu
,
F.
Ewald
,
A.
Fölster
,
A.S.
Picksak
,
S.
Harder
,
A.T.
El Gammal
, et al
.
2015
.
COSMC knockdown mediated aberrant O-glycosylation promotes oncogenic properties in pancreatic cancer
.
Mol. Cancer
.
14
:
109
.
Holdbrooks
,
A.T.
,
C.M.
Britain
, and
S.L.
Bellis
.
2018
.
ST6Gal-I sialyltransferase promotes tumor necrosis factor (TNF)-mediated cancer cell survival via sialylation of the TNF receptor 1 (TNFR1) death receptor
.
J. Biol. Chem.
293
:
1610
1622
.
Holst
,
S.
,
A.I.
Belo
,
E.
Giovannetti
,
I.
Van Die
, and
M.
Wuhrer
.
2017
.
Profiling of different pancreatic cancer cells used as models for metastatic behaviour shows large variation in their N-glycosylation
.
Sci. Rep.
7
:
16623
.
Houghton
,
J.L.
,
D.
Abdel-Atti
,
W.W.
Scholz
, and
J.S.
Lewis
.
2017
.
Preloading with unlabeled CA19.9 targeted human monoclonal antibody leads to improved PET imaging with (89)Zr-5B1
.
Mol. Pharm.
14
:
908
915
.
Hsieh
,
C.C.
,
Y.M.
Shyr
,
W.Y.
Liao
,
T.H.
Chen
,
S.E.
Wang
,
P.C.
Lu
,
P.Y.
Lin
,
Y.B.
Chen
,
W.Y.
Mao
,
H.Y.
Han
, et al
.
2017
.
Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructoseresponsive manner promotes pancreatic cancer metastasis
.
Oncotarget
.
8
:
7691
7709
.
Hsu
,
J.M.
,
C.W.
Li
,
Y.J.
Lai
, and
M.C.
Hung
.
2018
.
Posttranslational modifications of PD-L1 and their applications in cancer therapy
.
Cancer Res.
78
:
6349
6353
.
Itzkowitz
,
S.
,
T.
Kjeldsen
,
A.
Friera
,
S.
Hakomori
,
U.S.
Yang
, and
Y.S.
Kim
.
1991
.
Expression of Tn, sialosyl Tn, and T antigens in human pancreas
.
Gastroenterology
.
100
:
1691
1700
.
Iwai
,
K.
,
H.
Ishikura
,
M.
Kaji
,
H.
Sugiura
,
A.
Ishizu
,
C.
Takahashi
,
H.
Kato
,
T.
Tanabe
, and
T.
Yoshiki
.
1993
.
Importance of E-selectin (ELAM-1) and sialyl Lewis(a) in the adhesion of pancreatic carcinoma cells to activated endothelium
.
Int. J. Cancer
.
54
:
972
977
.
Jacobetz
,
M.A.
,
D.S.
Chan
,
A.
Neesse
,
T.E.
Bapiro
,
N.
Cook
,
K.K.
Frese
,
C.
Feig
,
T.
Nakagawa
,
M.E.
Caldwell
,
H.I.
Zecchini
, et al
.
2013
.
Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer
.
Gut
.
62
:
112
120
.
Jahan
,
R.
,
M.A.
Macha
,
S.
Rachagani
,
S.
Das
,
L.M.
Smith
,
S.
Kaur
, and
S.K.
Batra
.
2018
.
Axed MUC4 (MUC4/X) aggravates pancreatic malignant phenotype by activating integrin-β1/FAK/ERK pathway
.
Biochim. Biophys. Acta Mol. Basis Dis.
1864
:
2538
2549
.
Jayanth
,
P.
,
S.R.
Amith
,
K.
Gee
, and
M.R.
Szewczuk
.
2010
.
Neu1 sialidase and matrix metalloproteinase-9 cross-talk is essential for neurotrophin activation of Trk receptors and cellular signaling
.
Cell Signal
.
22
:
1193
1205
.
Jia
,
C.
,
H.
Li
,
D.
Fu
, and
Y.
Lan
.
2020
.
GFAT1/HBP/O-GlcNAcylation axis regulates β-catenin activity to promote pancreatic cancer aggressiveness
.
Biomed. Res. Int.
2020
:
1921609
.
Jonckheere
,
N.
, and
I.
Van Seuningen
.
2018
.
Integrative analysis of the cancer genome atlas and cancer cell lines encyclopedia large-scale genomic databases: MUC4/MUC16/MUC20 signature is associated with poor survival in human carcinomas
.
J. Transl Med.
16
:
259
.
Jones
,
R.B.
,
K.A.
Dorsett
,
A.B.
Hjelmeland
, and
S.L.
Bellis
.
2018
.
The ST6Gal-I sialyltransferase protects tumor cells against hypoxia by enhancing HIF-1α signaling
.
J. Biol. Chem.
293
:
5659
5667
.
Kaji
,
M.
,
H.
Ishikura
,
T.
Kishimoto
,
M.
Omi
,
A.
Ishizu
,
C.
Kimura
,
T.
Takahashi
,
H.
Kato
, and
T.
Yoshiki
.
1995
.
E-selectin expression induced by pancreas-carcinoma-derived interleukin-1 alpha results in enhanced adhesion of pancreas-carcinoma cells to endothelial cells
.
Int. J. Cancer
.
60
:
712
717
.
Kalra
,
A.V.
, and
R.B.
Campbell
.
2007
.
Mucin impedes cytotoxic effect of 5-FU against growth of human pancreatic cancer cells: Overcoming cellular barriers for therapeutic gain
.
Br. J. Cancer
.
97
:
910
918
.
Kalra
,
A.V.
, and
R.B.
Campbell
.
2009
.
Mucin overexpression limits the effectiveness of 5-FU by reducing intracellular drug uptake and antineoplastic drug effects in pancreatic tumours
.
Eur. J. Cancer
.
45
:
164
173
.
Kalthoff
,
H.
,
C.
Kreiker
,
W.H.
Schmiegel
,
H.
Greten
, and
H.G.
Thiele
.
1986
.
Characterization of CA 19-9 bearing mucins as physiological exocrine pancreatic secretion products
.
Cancer Res.
46
:
3605
3607
Kang
,
H.
,
Q.
Wu
,
A.
Sun
,
X.
Liu
,
Y.
Fan
, and
X.
Deng
.
2018
.
Cancer cell glycocalyx and its significance in cancer progression
.
Int. J. Mol. Sci.
19
:
E2484
.
Karsten
,
U.
,
C.
Diotel
,
G.
Klich
,
H.
Paulsen
,
S.
Goletz
,
S.
Müller
, and
F.G.
Hanisch
.
1998
.
Enhanced binding of antibodies to the DTR motif of MUC1 tandem repeat peptide is mediated by site-specific glycosylation
.
Cancer Res.
58
:
2541
2549
Karsten
,
U.
,
N.
Serttas
,
H.
Paulsen
,
A.
Danielczyk
, and
S.
Goletz
.
2004
.
Binding patterns of DTR-specific antibodies reveal a glycosylation-conditioned tumor-specific epitope of the epithelial mucin (MUC1)
.
Glycobiology
.
14
:
681
692
.
Kim
,
G.E.
,
H.I.
Bae
,
H.U.
Park
,
S.F.
Kuan
,
S.C.
Crawley
,
J.J.L.
Ho
, and
Y.S.
Kim
.
2002
.
Aberrant expression of MUC5AC and MUC6 gastric mucins and sialyl Tn antigen in intraepithelial neoplasms of the pancreas
.
Gastroenterology
.
123
:
1052
1060
.
Kleeff
,
J.
,
T.
Ishiwata
,
A.
Kumbasar
,
H.
Friess
,
M.W.
Buchler
,
A.D.
Lander
, and
M.
Korc
.
1998
.
The cell-surface heparan sulfate proteoglycan glypican-1 regulates growth factor action in pancreatic carcinoma cells and is overexpressed in human pancreatic cancer
.
J. Clin. Invest.
102
:
1662
1673
.
Kleeff
,
J.
,
M.
Korc
,
M.
Apte
,
C.
La Vecchia
,
C.D.
Johnson
,
A.V.
Biankin
,
R.E.
Neale
,
M.
Tempero
,
D.A.
Tuveson
,
R.H.
Hruban
, and
J.P.
Neoptolemos
.
2016
.
Pancreatic cancer
.
Nat. Rev. Dis. Primers
.
2
:
16022
.
Kohlgraf
,
K.G.
,
A.J.
Gawron
,
M.
Higashi
,
J.L.
Meza
,
M.D.
Burdick
,
S.
Kitajima
,
D.L.
Kelly
,
T.C.
Caffrey
, and
M.A.
Hollingsworth
.
2003
.
Contribution of the MUC1 tandem repeat and cytoplasmic tail to invasive and metastatic properties of a pancreatic cancer cell line
.
Cancer Res.
63
:
5011
5020
Kudelka
,
M.R.
,
T.
Ju
,
J.
Heimburg-Molinaro
, and
R.D.
Cummings
.
2015
.
Simple sugars to complex disease–mucin-type O-glycans in cancer
.
Adv. Cancer Res.
126
:
53
135
.
Kuo
,
T.C.
,
M.H.
Wu
,
S.H.
Yang
,
S.T.
Chen
,
T.W.
Hsu
,
J.Y.
Jhuang
,
Y.Y.
Liao
,
Y.W.
Tien
, and
M.C.
Huang
.
2021a
.
C1GALT1 high expression is associated with poor survival of patients with pancreatic ductal adenocarcinoma and promotes cell invasiveness through integrin αv
.
Oncogene
.
40
:
1242
1254
.
Kuo
,
T.C.
,
M.H.
Wu
,
S.H.
Yang
,
S.T.
Chen
,
T.W.
Hsu
,
J.Y.
Jhuang
,
Y.Y.
Liao
,
Y.W.
Tien
, and
M.C.
Huang
.
2021b
.
C1GALT1 high expression is associated with poor survival of patients with pancreatic ductal adenocarcinoma and promotes cell invasiveness through integrin α(v)
.
Oncogene
.
40
:
1242
1254
.
Li
,
C.W.
,
S.O.
Lim
,
E.M.
Chung
,
Y.S.
Kim
,
A.H.
Park
,
J.
Yao
,
J.H.
Cha
,
W.
Xia
,
L.C.
Chan
,
T.
Kim
, et al
.
2018
.
Eradication of triple-negative breast cancer cells by targeting glycosylated PD-L1
.
Cancer Cell
.
33
:
187
201.e10
.
Li
,
C.W.
,
S.O.
Lim
,
W.
Xia
,
H.H.
Lee
,
L.C.
Chan
,
C.W.
Kuo
,
K.H.
Khoo
,
S.S.
Chang
,
J.H.
Cha
,
T.
Kim
, et al
.
2016
.
Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity
.
Nat. Commun.
7
:
12632
.
Liu
,
Y.C.
,
H.Y.
Yen
,
C.Y.
Chen
,
C.H.
Chen
,
P.F.
Cheng
,
Y.H.
Juan
,
C.H.
Chen
,
K.H.
Khoo
,
C.J.
Yu
,
P.C.
Yang
, et al
.
2011
.
Sialylation and fucosylation of epidermal growth factor receptor suppress its dimerization and activation in lung cancer cells
.
Proc. Natl. Acad. Sci. USA
.
108
:
11332
11337
.
Lohrmann
,
C.
,
E.M.
O’reilly
,
J.A.
O’donoghue
,
N.
Pandit-Taskar
,
J.A.
Carrasquillo
,
S.K.
Lyashchenko
,
S.
Ruan
,
R.
Teng
,
W.
Scholz
,
P.W.
Maffuid
, et al
.
2019
.
Retooling a blood-based biomarker: Phase I assessment of the high-affinity CA19-9 antibody HuMab-5B1 for immuno-PET imaging of pancreatic cancer
.
Clin. Cancer Res.
25
:
7014
7023
.
Lyubsky
,
S.
,
J.
Madariaga
,
M.
Lozowski
,
Y.
Mishriki
,
A.
Schuss
,
S.
Chao
, and
J.
Lundy
.
1988
.
A tumor-associated antigen in carcinoma of the pancreas defined by monoclonal antibody B72.3
.
Am. J. Clin. Pathol.
89
:
160
167
.
Madsen
,
C.B.
,
K.
Lavrsen
,
C.
Steentoft
,
M.B.
Vester-Christensen
,
H.
Clausen
,
H.H.
Wandall
, and
A.E.
Pedersen
.
2013
.
Glycan elongation beyond the mucin associated Tn antigen protects tumor cells from immune-mediated killing
.
PLoS One
.
8
:e72413.
Mas
,
E.
,
C.
Crotte
,
D.
Lecestre
,
J.C.
Michalski
,
M.J.
Escribano
,
D.
Lombardo
, and
M.O.
Sadoulet
.
1997
.
The oncofetal J28 epitope involves fucosylated O-linked oligosaccharide structures of the fetoacinar pancreatic protein
.
Glycobiology
.
7
:
745
752
.
Mathew
,
M.P.
,
E.
Tan
,
C.T.
Saeui
,
P.
Bovonratwet
,
L.
Liu
,
R.
Bhattacharya
, and
K.J.
Yarema
.
2015
.
Metabolic glycoengineering sensitizes drug-resistant pancreatic cancer cells to tyrosine kinase inhibitors erlotinib and gefitinib
.
Bioorg. Med. Chem. Lett.
25
:
1223
1227
.
Mathew
,
M.P.
,
E.
Tan
,
C.T.
Saeui
,
P.
Bovonratwet
,
S.
Sklar
,
R.
Bhattacharya
, and
K.J.
Yarema
.
2016
.
Metabolic flux-driven sialylation alters internalization, recycling, and drug sensitivity of the epidermal growth factor receptor (EGFR) in SW1990 pancreatic cancer cells
.
Oncotarget
.
7
:
66491
66511
.
Mcdermott
,
K.M.
,
P.R.
Crocker
,
A.
Harris
,
M.D.
Burdick
,
Y.
Hinoda
,
T.
Hayashi
,
K.
Imai
, and
M.A.
Hollingsworth
.
2001
.
Overexpression of MUC1 reconfigures the binding properties of tumor cells
.
Int. J. Cancer
.
94
:
783
791
.
Möckl
,
L.
2020
.
The emerging role of the mammalian glycocalyx in functional membrane organization and immune system regulation
.
Front. Cell Dev. Biol.
8
:
253
.
Muniyan
,
S.
,
D.
Haridas
,
S.
Chugh
,
S.
Rachagani
,
I.
Lakshmanan
,
S.
Gupta
,
P.
Seshacharyulu
,
L.M.
Smith
,
M.P.
Ponnusamy
, and
S.K.
Batra
.
2016
.
MUC16 contributes to the metastasis of pancreatic ductal adenocarcinoma through focal adhesion mediated signaling mechanism
.
Genes Cancer
.
7
:
110
124
.
Munkley
,
J.
, and
D.J.
Elliott
.
2016
.
Hallmarks of glycosylation in cancer
.
Oncotarget
.
7
:
35478
35489
.
Nagarajan
,
A.
,
P.
Malvi
, and
N.
Wajapeyee
.
2018
.
Heparan sulfate and heparan sulfate proteoglycans in cancer initiation and progression
.
Front. Endocrinol.
9
:
483
.
Nagata
,
K.
,
M.
Horinouchi
,
M.
Saitou
,
M.
Higashi
,
M.
Nomoto
,
M.
Goto
, and
S.
Yonezawa
.
2007
.
Mucin expression profile in pancreatic cancer and the precursor lesions
.
J. Hepatobiliary Pancreat. Surg.
14
:
243
254
.
Nagel
,
A.K.
, and
L.E.
Ball
.
2015
.
Intracellular protein O-GlcNAc modification integrates nutrient status with transcriptional and metabolic regulation
.
Adv. Cancer Res.
126
:
137
166
.
Nath
,
D.
,
A.
Hartnell
,
L.
Happerfield
,
D.W.
Miles
,
J.
Burchell
,
J.
Taylor-Papadimitriou
, and
P.R.
Crocker
.
1999
.
Macrophage-tumour cell interactions: Identification of MUC1 on breast cancer cells as a potential counter-receptor for the macrophage-restricted receptor, sialoadhesin
.
Immunology
.
98
:
213
219
.
Ohtsubo
,
K.
, and
J.D.
Marth
.
2006
.
Glycosylation in cellular mechanisms of health and disease
.
Cell
.
126
:
855
867
.
Ohya
,
A.
,
K.
Yamanoi
,
H.
Shimojo
,
C.
Fujii
, and
J.
Nakayama
.
2017
.
Gastric gland mucin-specific O-glycan expression decreases with tumor progression from precursor lesions to pancreatic cancer
.
Cancer Sci.
108
:
1897
1902
.
Palucka
,
K.
, and
J.
Banchereau
.
2013
.
Dendritic-cell-based therapeutic cancer vaccines
.
Immunity
.
39
:
38
48
.
Pan
,
S.
,
R.
Chen
,
Y.
Tamura
,
D.A.
Crispin
,
L.A.
Lai
,
D.H.
May
,
M.W.
Mcintosh
,
D.R.
Goodlett
, and
T.A.
Brentnall
.
2014
.
Quantitative glycoproteomics analysis reveals changes in N-glycosylation level associated with pancreatic ductal adenocarcinoma
.
J. Proteome Res.
13
:
1293
1306
.
Pan
,
S.
,
Y.
Tamura
,
R.
Chen
,
D.
May
,
M.W.
Mcintosh
, and
T.A.
Brentnall
.
2012
.
Large-scale quantitative glycoproteomics analysis of site-specific glycosylation occupancy
.
Mol. Biosyst.
8
:
2850
2856
.
Peng
,
C.
,
Y.
Zhu
,
W.
Zhang
,
Q.
Liao
,
Y.
Chen
,
X.
Zhao
,
Q.
Guo
,
P.
Shen
,
B.
Zhen
,
X.
Qian
, et al
.
2017
.
Regulation of the Hippo-YAP pathway by glucose sensor O-GlcNAcylation
.
Mol. Cell
.
68
:
591
604.e5
.
Pérez-Garay
,
M.
,
B.
Arteta
,
E.
Llop
,
L.
Cobler
,
L.
Pagès
,
R.
Ortiz
,
M.J.
Ferri
,
C.
De Bolós
,
J.
Figueras
,
R.
De Llorens
, et al
.
2013
.
α2,3-Sialyltransferase ST3Gal IV promotes migration and metastasis in pancreatic adenocarcinoma cells and tends to be highly expressed in pancreatic adenocarcinoma tissues
.
Int. J. Biochem. Cell Biol.
45
:
1748
1757
.
Pérez-Garay
,
M.
,
B.
Arteta
,
L.
Pagès
,
R.
De Llorens
,
C.
De Bolòs
,
F.
Vidal-Vanaclocha
, and
R.
Peracaula
.
2010
.
alpha2,3-sialyltransferase ST3Gal III modulates pancreatic cancer cell motility and adhesion in vitro and enhances its metastatic potential in vivo
.
PLoS One
.
5
:e12524.
Phillips
,
M.L.
,
E.
Nudelman
,
F.C.
Gaeta
,
M.
Perez
,
A.K.
Singhal
,
S.
Hakomori
, and
J.C.
Paulson
.
1990
.
ELAM-1 mediates cell adhesion by recognition of a carbohydrate ligand, sialyl-Lex
.
Science
.
250
:
1130
1132
.
Pour
,
P.M.
,
M.M.
Tempero
,
H.
Takasaki
,
E.
Uchida
,
Y.
Takiyama
,
D.A.
Burnett
, and
Z.
Steplewski
.
1988
.
Expression of blood group-related antigens ABH, Lewis A, Lewis B, Lewis X, Lewis Y, and CA 19-9 in pancreatic cancer cells in comparison with the patient’s blood group type
.
Cancer Res.
48
:
5422
5426
Provenzano
,
P.P.
,
C.
Cuevas
,
A.E.
Chang
,
V.K.
Goel
,
D.D.
Von Hoff
, and
S.R.
Hingorani
.
2012
.
Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma
.
Cancer Cell
.
21
:
418
429
.
Provenzano
,
P.P.
, and
S.R.
Hingorani
.
2013
.
Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer
.
Br. J. Cancer
.
108
:
1
8
.
Qin
,
W.
,
L.
Hu
,
X.
Zhang
,
S.
Jiang
,
J.
Li
,
Z.
Zhang
, and
X.
Wang
.
2019
.
The diverse function of PD-1/PD-L pathway beyond cancer
.
Front. Immunol.
10
:
2298
.
Qorri
,
B.
,
W.
Harless
, and
M.R.
Szewczuk
.
2020
.
Novel molecular mechanism of aspirin and celecoxib targeting mammalian neuraminidase-1 impedes epidermal growth factor receptor signaling axis and induces apoptosis in pancreatic cancer cells
.
Drug Des. Devel. Ther.
14
:
4149
4167
.
Radhakrishnan
,
P.
,
S.
Dabelsteen
,
F.B.
Madsen
,
C.
Francavilla
,
K.L.
Kopp
,
C.
Steentoft
,
S.Y.
Vakhrushev
,
J.V.
Olsen
,
L.
Hansen
,
E.P.
Bennett
, et al
.
2014
.
Immature truncated O-glycophenotype of cancer directly induces oncogenic features
.
Proc. Natl. Acad. Sci. USA
.
111
:
E4066
E4075
.
Reis
,
C.A.
,
L.
David
,
M.
Seixas
,
J.
Burchell
, and
M.
Sobrinho-Simões
.
1998
.
Expression of fully and under-glycosylated forms of MUC1 mucin in gastric carcinoma
.
Int. J. Cancer
.
79
:
402
410
.
Remmers
,
N.
,
J.M.
Anderson
,
E.M.
Linde
,
D.J.
Dimaio
,
A.J.
Lazenby
,
H.H.
Wandall
,
U.
Mandel
,
H.
Clausen
,
F.
Yu
, and
M.A.
Hollingsworth
.
2013
.
Aberrant expression of mucin core proteins and o-linked glycans associated with progression of pancreatic cancer
.
Clin. Cancer Res.
19
:
1981
1993
.
Ringel
,
J.
, and
M.
Löhr
.
2003
.
The MUC gene family: Their role in diagnosis and early detection of pancreatic cancer
.
Mol. Cancer
.
2
:
9
Rodriguez
,
E.
,
K.
Boelaars
,
K.
Brown
,
Eveline
Li
,
R.J.
,
L.
Kruijssen
,
S.C.M.
Bruijns
,
T.
Van Ee
,
S.T.T.
Schetters
,
M.H.W.
Crommentuijn
, et al
.
2021
.
Sialic acids in pancreatic cancer cells drive tumour-associated macrophage differentiation via the Siglec receptors Siglec-7 and Siglec-9
.
Nat. Commun.
12
:
1270
.
Rowson-Hodel
,
A.R.
,
J.H.
Wald
,
J.
Hatakeyama
,
W.K.
O’neal
,
J.R.
Stonebraker
,
K.
Vandervorst
,
M.J.
Saldana
,
A.D.
Borowsky
,
C.
Sweeney
, and
K.L.
Carraway
.
2018
.
Membrane Mucin Muc4 promotes blood cell association with tumor cells and mediates efficient metastasis in a mouse model of breast cancer
.
Oncogene
.
37
:
197
207
.
Saeland
,
E.
,
Van
Vliet
,
S.J.
,
M.
Bäckström
,
V.C.
Van Den Berg
,
T.B.
Geijtenbeek
,
G.A.
Meijer
, and
Y.
Van Kooyk
.
2007
.
The C-type lectin MGL expressed by dendritic cells detects glycan changes on MUC1 in colon carcinoma
.
Cancer Immunol. Immunother.
56
:
1225
1236
.
Saleh
,
F.M.
,
P.K.
Chandra
,
D.
Lin
,
J.E.
Robinson
,
R.
Izadpanah
,
D.
Mondal
,
C.
Bollensdorff
,
E.U.
Alt
,
Q.
Zhu
,
W.A.
Marasco
, et al
.
2020
.
A new humanized mouse model mimics humans in lacking α-Gal epitopes and secreting anti-gal antibodies
.
J. Immunol.
204
:
1998
2005
.
Salnikov
,
A.V.
,
N.P.
Bretz
,
C.
Perne
,
J.
Hazin
,
S.
Keller
,
M.
Fogel
,
I.
Herr
,
T.
Schlange
,
G.
Moldenhauer
, and
P.
Altevogt
.
2013
.
Antibody targeting of CD24 efficiently retards growth and influences cytokine milieu in experimental carcinomas
.
Br. J. Cancer
.
108
:
1449
1459
.
Sato
,
N.
,
S.
Kohi
,
K.
Hirata
, and
M.
Goggins
.
2016
.
Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight
.
Cancer Sci.
107
:
569
575
.
Satoh
,
S.
,
Y.
Hinoda
,
T.
Hayashi
,
M.D.
Burdick
,
K.
Imai
, and
M.A.
Hollingsworth
.
2000
.
Enhancement of metastatic properties of pancreatic cancer cells by MUC1 gene encoding an anti-adhesion molecule
.
Int. J. Cancer
.
88
:
507
518
.
Sawada
,
T.
,
J.J.
Ho
,
Y.S.
Chung
,
M.
Sowa
, and
Y.S.
Kim
.
1994
.
E-selectin binding by pancreatic tumor cells is inhibited by cancer sera
.
Int. J. Cancer
.
57
:
901
907
.
Sawada
,
T.
,
J.J.
Ho
,
T.
Sagabe
,
W.H.
Yoon
,
Y.S.
Chung
,
M.
Sowa
, and
Y.S.
Kim
.
1993
.
Biphasic effect of cell surface sialic acids on pancreatic cancer cell adhesiveness
.
Biochem. Biophys. Res. Commun.
195
:
1096
1103
.
Schultz
,
M.J.
,
A.T.
Holdbrooks
,
A.
Chakraborty
,
W.E.
Grizzle
,
C.N.
Landen
,
D.J.
Buchsbaum
,
M.G.
Conner
,
R.C.
Arend
,
K.J.
Yoon
,
C.A.
Klug
, et al
.
2016
.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype
.
Cancer Res.
76
:
3978
3988
.
Sharma
,
N.S.
,
V.K.
Gupta
,
P.
Dauer
,
K.
Kesh
,
R.
Hadad
,
B.
Giri
,
A.
Chandra
,
V.
Dudeja
,
C.
Slawson
,
S.
Banerjee
, et al
.
2019
.
O-GlcNAc modification of Sox2 regulates self-renewal in pancreatic cancer by promoting its stability
.
Theranostics
.
9
:
3410
3424
.
Sharma
,
P.
, and
J.P.
Allison
.
2015
.
Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential
.
Cell
.
161
:
205
214
.
Shin
,
H.
,
H.J.
Cha
,
K.
Na
,
M.J.
Lee
,
J.Y.
Cho
,
C.Y.
Kim
,
E.K.
Kim
,
C.M.
Kang
,
H.
Kim
, and
Y.K.
Paik
.
2018
.
O-GlcNAcylation of the tumor suppressor FOXO3 triggers aberrant cancer cell growth
.
Cancer Res.
78
:
1214
1224
.
Singh
,
P.K.
,
Y.
Wen
,
B.J.
Swanson
,
K.
Shanmugam
,
A.
Kazlauskas
,
R.L.
Cerny
,
S.J.
Gendler
, and
M.A.
Hollingsworth
.
2007
.
Platelet-derived growth factor receptor beta-mediated phosphorylation of MUC1 enhances invasiveness in pancreatic adenocarcinoma cells
.
Cancer Res.
67
:
5201
5210
.
Singh
,
S.
,
K.
Pal
,
J.
Yadav
,
H.
Tang
,
K.
Partyka
,
D.
Kletter
,
P.
Hsueh
,
E.
Ensink
,
B.
Kc
,
G.
Hostetter
, et al
.
2015
.
Upregulation of glycans containing 3′ fucose in a subset of pancreatic cancers uncovered using fusion-tagged lectins
.
J. Proteome Res.
14
:
2594
2605
.
Stanley
,
P.
2011
.
Golgi glycosylation
.
Cold Spring Harb Perspect. Biol.
3
:
a005199
.
Suh
,
H.
,
K.
Pillai
, and
D.L.
Morris
.
2017
.
Mucins in pancreatic cancer: Biological role, implications in carcinogenesis and applications in diagnosis and therapy
.
Am. J. Cancer Res.
7
:
1372
1383
Sutherlin
,
M.E.
,
I.
Nishimori
,
T.
Caffrey
,
E.P.
Bennett
,
H.
Hassan
,
U.
Mandel
,
D.
Mack
,
T.
Iwamura
,
H.
Clausen
, and
M.A.
Hollingsworth
.
1997
.
Expression of three UDP-N-acetyl-alpha-D-galactosamine:polypeptide GalNAc N-acetylgalactosaminyltransferases in adenocarcinoma cell lines
.
Cancer Res.
57
:
4744
4748
Swanson
,
B.J.
,
K.M.
Mcdermott
,
P.K.
Singh
,
J.P.
Eggers
,
P.R.
Crocker
, and
M.A.
Hollingsworth
.
2007
.
MUC1 is a counter-receptor for myelin-associated glycoprotein (Siglec-4a) and their interaction contributes to adhesion in pancreatic cancer perineural invasion
.
Cancer Res.
67
:
10222
10229
.
Takada
,
A.
,
K.
Ohmori
,
N.
Takahashi
,
K.
Tsuyuoka
,
A.
Yago
,
K.
Zenita
,
A.
Hasegawa
, and
R.
Kannagi
.
1991
.
Adhesion of human cancer cells to vascular endothelium mediated by a carbohydrate antigen, sialyl Lewis A
.
Biochem. Biophys. Res. Commun.
179
:
713
719
.
Takada
,
A.
,
K.
Ohmori
,
T.
Yoneda
,
K.
Tsuyuoka
,
A.
Hasegawa
,
M.
Kiso
, and
R.
Kannagi
.
1993
.
Contribution of carbohydrate antigens sialyl Lewis A and sialyl Lewis X to adhesion of human cancer cells to vascular endothelium
.
Cancer Res.
53
:
354
361
Takada
,
M.
,
M.
Yamamoto
,
Y.
Hasegawa
, and
Y.
Saitoh
.
1995
.
Endothelial leukocyte adhesion molecule-1-mediated vasoinvasion of human pancreatic adenocarcinoma
.
J. Surg. Res.
59
:
653
657
.
Tang
,
H.
,
S.
Singh
,
K.
Partyka
,
D.
Kletter
,
P.
Hsueh
,
J.
Yadav
,
E.
Ensink
,
M.
Bern
,
G.
Hostetter
,
D.
Hartman
, et al
.
2015
.
Glycan motif profiling reveals plasma sialyl-lewis x elevations in pancreatic cancers that are negative for sialyl-lewis A
.
Mol. Cell Proteomics
.
14
:
1323
1333
.
Taniguchi
,
N.
, and
Y.
Kizuka
.
2015
.
Glycans and cancer: Role of N-glycans in cancer biomarker, progression and metastasis, and therapeutics
.
Adv. Cancer Res.
126
:
11
51
.
Taniuchi
,
K.
,
R.L.
Cerny
,
A.
Tanouchi
,
K.
Kohno
,
N.
Kotani
,
K.
Honke
,
T.
Saibara
, and
M.A.
Hollingsworth
.
2011
.
Overexpression of GalNAc-transferase GalNAc-T3 promotes pancreatic cancer cell growth
.
Oncogene
.
30
:
4843
4854
.
Tarp
,
M.A.
, and
H.
Clausen
.
2008
.
Mucin-type O-glycosylation and its potential use in drug and vaccine development
.
Biochim. Biophys. Acta.
1780
:
546
563
.
Teranishi
,
F.
,
N.
Takahashi
,
N.
Gao
,
Y.
Akamo
,
H.
Takeyama
,
T.
Manabe
, and
T.
Okamoto
.
2009
.
Phosphoinositide 3-kinase inhibitor (wortmannin) inhibits pancreatic cancer cell motility and migration induced by hyaluronan in vitro and peritoneal metastasis in vivo
.
Cancer Sci.
100
:
770
777
.
Thomas
,
D.
,
S.
Sagar
,
X.
Liu
,
H.R.
Lee
,
J.A.
Grunkemeyer
,
P.M.
Grandgenett
,
T.
Caffrey
,
K.A.
O’connell
,
B.
Swanson
,
L.
Marcos-Silva
, et al
.
2021
.
Isoforms of MUC16 activate oncogenic signaling through EGF receptors to enhance the progression of pancreatic cancer
.
Mol. Ther.
29
:
1557
1571
.
Tinder
,
T.L.
,
D.B.
Subramani
,
G.D.
Basu
,
J.M.
Bradley
,
J.
Schettini
,
A.
Million
,
T.
Skaar
, and
P.
Mukherjee
.
2008
.
MUC1 enhances tumor progression and contributes toward immunosuppression in a mouse model of spontaneous pancreatic adenocarcinoma
.
J. Immunol.
181
:
3116
3125
.
Topalian
,
S.L.
,
C.G.
Drake
, and
D.M.
Pardoll
.
2015
.
Immune checkpoint blockade: A common denominator approach to cancer therapy
.
Cancer Cell
.
27
:
450
461
.
Topalian
,
S.L.
,
J.M.
Taube
,
R.A.
Anders
, and
D.M.
Pardoll
.
2016
.
Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy
.
Nat. Rev. Cancer
.
16
:
275
287
.
Uchimido
,
R.
,
E.P.
Schmidt
, and
N.I.
Shapiro
.
2019
.
The glycocalyx: A novel diagnostic and therapeutic target in sepsis
.
Crit. Care
.
23
:
16
.
Varki
,
A.
,
R.D.
Cummings
,
J.D.
Esko
,
P.
Stanley
,
G.W.
Hart
,
M.
Aebi
,
A.G.
Darvill
,
T.
Kinoshita
,
N.H.
Packer
,
J.H.
Prestegard
, et al
.
2015
.
Essentials of Glycobiology [Internet]
. Third edition.
Cold Spring Harbor
,
NY
.
Vedadghavami
,
A.
,
C.
Zhang
, and
A.G.
Bajpayee
.
2020
.
Overcoming negatively charged tissue barriers: Drug delivery using cationic peptides and proteins
.
Nano Today
.
34
:
100898
.
Vennin
,
C.
,
P.
Melenec
,
R.
Rouet
,
M.
Nobis
,
A.S.
Cazet
,
K.J.
Murphy
,
D.
Herrmann
,
D.A.
Reed
,
M.C.
Lucas
,
S.C.
Warren
, et al
.
2019
.
CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan
.
Nat. Commun.
10
:
3637
.
Wang
,
Y.
,
H.
Wang
,
H.
Yao
,
C.
Li
,
J.Y.
Fang
, and
J.
Xu
.
2018
.
Regulation of PD-L1: Emerging routes for targeting tumor immune evasion
.
Front. Pharmacol.
9
:
536
.
Weitzenfeld
,
P.
,
S.
Bournazos
, and
J.V.
Ravetch
.
2019
.
Antibodies targeting sialyl Lewis A mediate tumor clearance through distinct effector pathways
.
J. Clin. Invest.
129
:
3952
3962
.
Yamada
,
N.
,
T.
Hamada
,
M.
Goto
,
H.
Tsutsumida
,
M.
Higashi
,
M.
Nomoto
, and
S.
Yonezawa
.
2006
.
MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer
.
Int. J. Cancer
.
119
:
1850
1857
.
Yang
,
Y.
,
V.
Macleod
,
H.Q.
Miao
,
A.
Theus
,
F.
Zhan
,
J.D.J.R.
Shaughnessy
,
J.
Sawyer
,
J.P.
Li
,
E.
Zcharia
,
I.
Vlodavsky
, and
R.D.
Sanderson
.
2007
.
Heparanase enhances syndecan-1 shedding: A novel mechanism for stimulation of tumor growth and metastasis
.
J. Biol. Chem.
282
:
13326
13333
.
Yao
,
W.
,
J.L.
Rose
,
W.
Wang
,
S.
Seth
,
H.
Jiang
,
A.
Taguchi
,
J.
Liu
,
L.
Yan
,
A.
Kapoor
,
P.
Hou
, et al
.
2019
.
Syndecan 1 is a critical mediator of macropinocytosis in pancreatic cancer
.
Nature
.
568
:
410
414
.
Yonezawa
,
S.
,
A.
Nakamura
,
M.
Horinouchi
, and
E.
Sato
.
2002
.
The expression of several types of mucin is related to the biological behavior of pancreatic neoplasms
.
J. Hepatobiliary Pancreat. Surg.
9
:
328
341
.
Yonezawa
,
S.
, and
E.
Sato
.
1997
.
Expression of mucin antigens in human cancers and its relationship with malignancy potential
.
Pathol. Int.
47
:
813
830
.
Zou
,
W.
,
J.D.
Wolchok
, and
L.
Chen
.
2016
.
PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations
.
Sci. Transl. Med.
8
:
328rv4
.

Author notes

Disclosures: The authors declare no competing interests exist.

This article is available under a Creative Commons License (Attribution 4.0 International, as described at https://creativecommons.org/licenses/by/4.0/).