You are on page 1of 10

BBA - Reviews on Cancer 1868 (2017) 157–166

Contents lists available at ScienceDirect

BBA - Reviews on Cancer


journal homepage: www.elsevier.com/locate/bbacan

Glycoconjugates from extracellular vesicles: Structures, functions and MARK


emerging potential as cancer biomarkers
Julia Costa
Laboratory of Glycobiology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal

A R T I C L E I N F O A B S T R A C T

Keywords: Extracellular vesicles (EVs) are released by virtually all cells, carry cellular molecules to the extracellular
Cancer biomarkers environment, and may interact with other cells. They are found in body fluids, therefore, constituting useful
Exosomes target sources for the identification of disease biomarkers, for example, in cancer. EVs originate from the plasma
Extracellular vesicles membrane or from multivesicular endosomes. They have the same topology as the plasma membrane and are
Glycoconjugates
rich in glycoconjugates, displaying specific glycosignatures. Surface glycoconjugates play important roles in EVs
Glycoproteins
Glycosylation
biogenesis and in their interaction with other cells. Changes in glycosylation constitute a hallmark of different
types of cancer, therefore, the study of glycoconjugates and glycosignatures of EVs appear as promising
candidates to identify novel cancer biomarkers and to increase the specificity and sensitivity of the existing
clinical biomarkers, many of which are glycosylated.

1. Introduction 2. Glycosylation

Cells release vesicles to their surroundings known as extracellular 2.1. Glycoconjugates


vesicles (EVs). EVs may originate from plasma membrane budding
(microvesicles) or from fusion of multivesicular endosomes with the In eukaryotic cells several classes of glycoconjugates can be found,
plasma membrane (exosomes) [1]. Increasing evidence has shown that which include glycoproteins and glycolipids. Glycosylation is a com-
EVs have a relevant functional role in transferring biomolecules from mon post-translational modification of proteins, N- and O-glycosylation
cell to cell, thereby constituting vehicles of communication among being the most widely studied. In N-glycosylation, glycans are cova-
cells. This is relevant in the transmission of pathogenic, signal and lently bound via a nitrogen atom to asparagine side chains of proteins
regulatory molecules in disease, such as cancer [1,2]. On the other (Fig. 1). In animal cells, the sugar linked to the asparagine residue is N-
hand, they can also transfer beneficial factors involved in regeneration acetylglucosamine (GlcNAc). N-glycosylation is initiated in the endo-
[3]. plasmic reticulum with the transfer of an oligosaccharide precursor
EVs are found in biological fluids and are capable of crossing onto the consensus acceptor sequence NXS/T in the nascent polypeptide
biological barriers [4]. Since they carry cellular material to the chain. This precursor is further processed by the action of glycosidases
extracellular environment, they are useful targets for the identification and glycosyltransferases in the endoplasmic reticulum and the Golgi
of disease biomarkers [5]. apparatus concomitant to the trafficking of proteins to the plasma
There is ample evidence in the literature showing that EVs are membrane, lysosome or the extracellular environment. There are
strongly glycosylated being rich in specific glycoconjugates [6–9]. In several classes of N-glycans: i) high mannose-type that only contain
this article we address the importance of glycoconjugates and their mannose (Man) and GlcNAc residues; ii) complex-type that also have
glycosylation in EVs properties and outline their emerging potential as other monosaccharide residues, e.g., galactose (Gal), N-acetylneurami-
cancer biomarkers. nic acid (NeuAc), fucose (Fuc); iii) hybrid-type in which one arm has
only Man and the other is processed to the complex type.
In O-glycosylation, glycans are bound via an oxygen atom to side
chains of serine, threonine, hydroxylysine or hydroxyproline residues.

Abbreviations: CA19-9, carbohydrate antigen 19-9; CEA, carcinoembryonic antigen; EV, extracellular vesicle; Fuc, fucose; Gal, galactose; GlcNAc, N-acetylglucosamine; GPI,
glycosylphosphatidylinositol; HER2, human epidermal growth factor receptor 2; HSPG, heparan sulfate proteoglycan; Man, mannose; MUC1, mucin 1; MUC16, mucin 16; NeuAc, N-
acetylneuraminic acid
E-mail address: jcosta@itqb.unl.pt.

http://dx.doi.org/10.1016/j.bbcan.2017.03.007
Received 3 February 2017; Received in revised form 20 March 2017; Accepted 21 March 2017
Available online 24 March 2017
0304-419X/ © 2017 Elsevier B.V. All rights reserved.
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

Fig. 1. Schematic representation of composition and topology of glycoconjugates in EVs.N-glycans of the complex and high mannose type are found in EVs [8]. HSPGs are present in EVs
and a schematic representation of the glycan moiety [10] is shown. Mucins are found in EVs and a representation of O-linked glycans commonly attached to mucins are presented [18]. O-
GlcNAcylated glycoconjugates are detected in EVs [90]. Glycans are represented according to the nomenclature of the Consortium for Functional Glycomics.

A major class of O-linked glycoproteins consists of mucins, where the golipids or glycophospholipids, respectively. Glycosphingolipids con-
sugar linked to serine or threonine residues is N-acetylgalactosamine tain a high diversity of structures including the most common ganglio-
(Fig. 1). In mucin O-glycosylation sequential addition of monosacchar- sides (e.g., sialylated GM3, GM2, GM1, GD1a, GD2) (Fig. 1). Glyco-
ide residues by glycosyltransferases occurs in the Golgi apparatus. sphingolipids may associate in lipid microdomains in the membrane
Mucins are heavily glycosylated and they may be attached to the that serve as important signaling platforms [11]. Glycolipids based on
membrane or be secreted. Another important class of O-glycosylated diacylglycerol that mediate the anchoring of proteins to the membrane
proteins consists of proteoglycans, where the carbohydrate moiety are designated as GPI-anchors (Fig. 1). GPI-anchored proteins are
(glycosaminoglycan) bound to the polypeptide chain (usually serine- predominantly localized on the plasma membrane generally in lipid
glycine sequences) is comparably very large and may have up to microdomains.
hundred monosaccharide residues. Proteoglycans may be attached to
the plasma membrane or be deposited in the extracellular matrix. The
2.2. Analytical techniques
attached glycans are linear and composed of disaccharides of alternat-
ing residues of amino sugar and hexose derivatives. Similar to mucins,
Glycans have more complex structures than other macromolecules
proteoglycan glycosylation involves the sequential addition of mono-
since they can be branched, there are different types of possible
saccharides by glycosyltransferases in the Golgi; these glycans are
glycosidic bonds and the linkage at the anomeric carbon can either
commonly sulfated in the Golgi concomitantly to biosynthesis, and
be α or β. Furthermore, glycoproteins with interesting biological roles
proteoglycans sulfation may be further regulated at the cell surface or
are often much less abundant than other cellular proteins. In view of
after secretion by the action of sulfatases [10]. There are different
this, the development of diversified and highly specific techniques for
groups of glycosaminoglycans based on monosaccharide composition
the analysis of glycan structures is required. The systematic study of
and sulfation: heparan sulfate and heparin, chondroitin sulfate and
glycosylation is approached by glycomics where total glycans (gly-
dermatan sulfate, hyaluronan and keratan sulfate. Syndecans and
come) are analysed, or by glycoproteomics where glycoprotein identi-
glypicans are heparan sulfate proteoglycans (HSPGs) that are bound
fication, sites of glycosylation and their occupancy including the
to the cell surface, the first consist of transmembrane proteoglycans
corresponding glycan structures are of relevance. Comprehensive re-
whereas the second are anchored to the membrane via a glycosylpho-
views on glycomics and glycoproteomics also with implications in the
sphatidylinositol (GPI)-anchor. HSPG is composed of disaccharides of D-
cancer biomarker field can be found in the literature [12–14].
glucuronic acid/L-iduronic acid and N-acetylglucosamine that are
In brief, for general characterization of glycosylation lectins can be
bound to the polypeptide chain via an O-xylose residue (Fig. 1).
used, which are proteins that bind to specific glycan structures. Lectins
Nuclear, cytoplasmic and mitochondrial proteins can also be modified
can be applied in blots [15,16], lectin arrays [6,7] or in lectin affinity
by addition of O-linked N-acetylglucosamine to serine or threonine
purification [17].
residues (Fig. 1).
Anti-glycan antibodies that recognize specific glycoepitopes are also
Another class of glycoconjugates found in cellular membranes are
used in immunoblots, immunohistochemistry and flow cytometry (e.g.,
glycolipids. Glycolipids may be embedded in the membrane via
anti-sialyl-Tn, anti-Lewis structures, anti-polysialic acid) [18], with
ceramide or via phosphatidylglycerol being designated as glycosphin-
strong applications in cancer diagnosis and prognosis.

158
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

Fig. 2. Diagrammatic representation of production of EVs and their interaction with recipient cells. Glycans from glycoconjugates are represented as hexagons.

For structure analysis N-glycans are in general released enzymati- bisecting GlcNAc has been found associated with ovarian cancer cells
cally with peptide N-glycosidase F from glycoproteins or derived and tissues [8,27,28].
glycopeptides [15], and also chemically by hydrazinolysis [19]. Con- Proximal fucosylation of N-glycans and the corresponding enzyme
cerning O-glycans, they are generally cleaved from the protein back- FUT8 are upregulated in several types of cancer including hepatocel-
bone in alkaline solution by β-elimination. To increase the sensitivity of lular carcinoma. As a consequence proximally fucosylated α-fetopro-
detection, glycans may be derivatized, for example, with 2-aminoben- tein, AFP-L3 appeared as a biomarker for liver cancer diagnosis [29].
zamide for fluorescence detection [8] or by permethylation [20] for Changes in O-glycosylation of the mucin type have also been
mass spectrometry. reported in cancer. Truncated structures have been observed with
The profiling and detailed structure analysis of glycans is usually increased levels of the T and Tn epitopes and sialylated derivatives
done by liquid chromatography and mass spectrometry techniques (e.g., sialyl-Tn) (Fig. 1) in different types of cancer including, breast,
including high/ultra performance liquid chromatography (H/UPLC), colorectal, gastric, pancreatic, lung and salivary gland cancer. The
HPAEC-PAD capillary electrophoresis and mass spectrometric (MS) potential of these altered structures for drug and vaccine development
techniques [8,15,21]. For determination of the type of linkage tandem has been discussed in the literature [18].
mass spectrometry (MS/MS) [22], endo- and exoglycosidase digestion Major changes in peripheral sialylation and fucosylation of glycans
[8,23] and/or methylation analysis [19] are performed. Although less are associated with cancer and are currently evaluated as markers for
sensitive NMR spectroscopy is also useful for the determination of improved diagnosis, prognosis and in response to treatment. The
glycan primary structure and elucidation of glycans conformation. This fucosylated and sialylated structures that compose the sialylated
later aspect may also be unveiled in some cases from X-ray crystal- Lewis determinants (Fig. 1) present in O-glycans and N-glycans
lography, although this is a challenging aspect due to the heterogeneity participate in cell adhesion events underlying the formation of metas-
and mobility of glycans at the surface of glycoproteins. tases via interaction with selectins [24]. Their synthesis involves the
The assignment of structures to specific glycosylation sites relies on action of α2,3-sialyltransferases and α1,3/4 fucosyltransferases
mass spectrometry analysis (LC/MS/MS) [13]. [19,30,31].
For the study of glycolipids mass spectrometry is commonly Glycosaminoglycans either O-linked to the polypeptide chain (pro-
performed. teoglycans) or free play different roles in physiology through their
In general, complementary techniques are used to establish the action on cell surface receptors and interaction with growth factors.
glycosylation profiles from glycoconjugates, cells or tissues including HSPGs in cancer cells are relevant for tumor growth, are associated
body fluids, with the aim of elucidating specific glycosignatures with bad prognosis and constitute targets for anti-cancer therapies [32].
relevant in given biological or physiological contexts. When consider- The levels of gangliosides are often deregulated in cancer. For
ing specific glycoproteins the establishment of glycosignatures includes example, the disialoganglioside GD2 was reported as marker for several
in addition to the repertoire of glycostructures (from the different cancers [33].
glycoforms) information about specific site occupancy. These issues are
highly relevant in context of potential cancer biomarker identification
(Section 4.3). 3. Extracellular vesicles

3.1. Characteristics and biogenesis


2.3. Changes of glycosylation in cancer
EVs are produced by many types of mammalian cells, including
Changes in glycosylation occur in many diseases and they have been tumor cells, cells of the immune system, neurons and glial cells. They
widely studied in cancer where neoglycoepitopes are detected asso- are designated as exosomes when they have an endosomal origin and
ciated with tumors and are used as disease biomarkers [18,24,25]. are produced from the fusion of multivesicular endosomes with the
Concerning N-glycosylation, most remarkable is the increased plasma membrane releasing the intraluminal vesicles to the extracel-
branching of N-glycans as the product of GlcNAc-transferase V, which lular environment (Fig. 2). In contrast, EVs that are produced from
is observed in different types of cancer [26]. On the other hand, plasma membrane budding are currently termed microvesicles. Exo-
GlcNAc-transferase III synthesizes bisecting-GlcNAc-containing N-gly- somes and microvesicles have the same topology as the plasma
cans, which usually prevents further branching of N-glycans. However, membrane. Exosomes have diameters below 150 nm whereas micro-

159
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

vesicles are more heterogeneous with sizes typically up to 1 μm [34]. EVs from different cells and body fluids contain specific glycocon-
However, the complexity is high and the classification of EVs is jugates with characteristic glycan structures (Table 1, Fig. 1). Initial
currently a challenge; supporting this notion, recently, several sub- studies showed the presence of distinct prion PrP glycoforms in
populations of EVs from dendritic cells were defined based on exosomes [41]. Furthermore, the enrichment in high mannose epitopes,
proteomic analysis [35]. In recent years, the literature on EVs has complex N-linked glycans, N-acetyllactosamine, sialic acid and fucosy-
greatly increased and in many papers the term “exosomes” has been lated epitopes concomitant to exclusion of blood group antigens A/B in
applied to a mixture of EVs from different cellular origins and with microvesicles were detected in microvesicles and HIV-1 from several T-
different properties. cell lines by lectin array technology [6]. In agreement, and using the
EVs have characteristic compositions in nucleic acids (mRNA, same strategy, specific glycosignatures were found in microvesicles of
miRNA), proteins, including glycoproteins, and lipids including glyco- T-cell lines, as well as skin cancer and colon cancer cell lines, with
lipids. Data concerning the composition of EVs from various sources are enrichment in high mannose, polylactosamine, α2,6-sialic acid, and
available in several databases: EVpedia (http://evpedia.info/) [36], complex N-glycans concomitant to depletion of terminal blood group A
Vesiclepedia (http://www.microvesicles.org) [37] and Exocarta and B antigens [7]. Lectin array technology allowed the detection of
(http://www.exocarta.org) [38]. glycan structures present in glycoconjugates from EVs surface. On the
Exosomes are formed after the invagination of the endosome other hand, in SKOV3 and OVMz ovarian carcinoma cells specific
membrane in a process assisted by the proteins from the endosomal glycoproteins and glycan profiles were also found for EVs using lectin
sorting complex required for transport (ESCRT)-0, -I, -II, -III and the blotting, which allowed the detection of N- and O-glycans linked to
associated proteins, which results in the formation of intraluminal glycoproteins [8,16,42]. Furthermore, complex N-glycans of the di-,
vesicles, and the appearance of multivesicular endosomes (also known tri-, and tetraantennary type with or without proximal fucose and
as multivesicular bodies based on electron microscopy observations). bisecting GlcNAc as well as high mannose glycans released with peptide
Several proteins that are used as exosome markers common to many N-glycosidase F from glycoproteins of SKOV3 cells were detected by
mammalian cells are involved in exosome biogenesis, for example, NP-HPLC complemented with mass spectrometry [8]. In addition,
Tsg101 from ESCRT-I or Alix. Proteins that are involved in membrane proteomic analysis allowed the identification of an abundant sialogly-
fusion and transport, such as RabGTPases, annexins or flotillin, are also coprotein as galectin-3 binding protein (LGALS3BP also known as Mac-
found in exosomes. When the endosome membrane invaginates several 2 binding protein) [8,16]; LGALS3BP was suggested as a potential
cytoplasmic proteins, such as heat-shock proteins (hsc70 and hsc90) or exosome marker.
cytoskeleton proteins, are included into the exosomes [34]. Whether N-glycoproteomic/glycomic analysis of urinary exosomes showed
there are specific mechanisms of enrichment of certain cytoplasmic the presence of complex, hybrid and high mannose-type N-glycans from
proteins is not clear at present. glycoproteins; glycans containing sialic acid-, bisecting GlcNAc and
EVs are enriched in tetraspanins, which are palmitoylated mem- peripheral as well as proximal Fuc were detected and evidence for
brane proteins that may be glycosylated and have four transmembrane sulfated and/or phosphorylated glycans was found [43]. In other
domains. They participate in protein-protein interactions, including studies, high mannose and sialylated complex glycans were detected
oligomerization and interaction with other proteins such as integrins, in urinary exovesicles [44,45] as well as GlcNAc-containing glycocon-
immunoglobulin-superfamily receptors and metalloproteinases. They jugates [46]. In galactosemia patients a shift from prevalent high
also interact with specific lipids such as cholesterol and gangliosides. mannose to complex glycans was observed. This shift was specific to
Tetraspanins like CD63, CD9, CD81 and CD82 are currently used as EVs EVs since it was not detected in the urinary Tamm-Horsfall glycoprotein
markers [39]. Tetraspanins are also involved in protein sorting into [44]. Therefore, EVs glycans constituted useful biomarkers in galacto-
exosomes, for example, in CD81-deficient cells exosomes are depleted semia, which is a potentially lethal hepatotoxic syndrome for new born
of CD81 ligands [40]. infants and that in the long-term causes complications related with the

Table 1
Selected glycoconjugates and glycans associated with EVs.

Molecule Biofluid/tumor cells Cancer/disease References

Prion Infected moRK13, GT1-7 cells Prion disease [41]


ADAM10 SKOV3 cells Ovarian [51]
Specific glycosignatures T-cells (Jurkat-Tat-CCR5, SupT1and H9) – [6]
Specific glycosignatures T-cells (Jurkat-Tat-CCR5, SupT1and H9) – [7]
HCT-15 and HT-29 cells Colon
SkMel-5 cells Melanoma
Breast milk –
Specific glycosignatures SKOV3 cells Ovarian [42]
H4 cells Neuroglioma
N-glycan profiles Urine Galactosemia [44]
EGFR, EGFRvIII, podoplanin SkMG3, GBM20/3, GLI36vIII, GLI36R132H, LNZ308, A172 cells and blood Glioblastoma [79]
Specific glycosignatures, LGALS3BP SKOV3, OVMz cells Ovarian [8,16]
Glycan profiles Urine – [45]
Syndecan-1,-2,-4, glypican U-87 MG and LN18 cells Glioblastoma [48]
a2,3-Linked sialic acid B cells – [73]
N-glycoproteins Urine – [43]
EWI-2 N-glycans Sk-Mel-5 cells Melanoma [58]
Highly glycosylated EMMPRIN MCF-7 cells Breast cancer [89]
CD24, EpCAM CaOV3, OVCAR3, SKOV3, OV90, OVCA429, UCI101, ES-2, TOV112D, TOV21G, UWB1.289, GG, M130, Ovarian [81,82]
ascites
GlcNAc-containing glycans Urine – [46]
Integrins Several cancer cells, tissues, blood Melanoma [65,67]
Breast
Pancreatic
O-GlcNAc SW480 and SW620 cells Colorectal [90]

160
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

central nervous system and ovaries. galectin-3 in melanoma cells [58]. Furthermore, galectin-5 has been
EVs contain heparan sulfate proteoglycans, and syndecans play a proposed to mediate the concentration of poly-N-lactosamine contain-
major role in exosome biogenesis (Section 4.1) and are relevant in the ing glycoproteins into reticulocytes [60]. It is quite known that glycan-
interaction between exosomes and target cells (Section 4.2). lectin interactions are involved in the sorting of glycoproteins in other
Concerning lipids EVs also have characteristic compositions and cellular settings [61]. Therefore, an alternative pathway of glycoprotein
they are enriched in glycosphingolipids (including gangliosides GM3, sorting to EVs that would involve glycans and lectins is not too far
GM1, GD2), sphingomyelin, cholesterol, and phosphatidylserine [47]. fetching. In this scenario glycans would interact with lectins and
On the other hand, GPI-anchored proteins are present in EVs, for promote the specific sorting into EVs possibly via glycoprotein oligo-
example, glypican [48]. merization or enrichment in membrane domains. More experimental
EVs contain nucleic acids including mRNA, miRNA and DNA and, evidence is still required in support of this hypothesis.
therefore, may serve as vehicles for the transfer of genetic information To proceed further in elucidating sorting mechanisms of specific
among cells and under disease conditions could be a source for the glycoconjugates into EVs, improved purification strategies to obtain
identification of biomarkers. This topic is subject of intensive research more homogeneous fractions of EVs, as well as plasma membrane and
[1]. multivesicular endosome membrane fractions will be helpful.

3.2. Separation techniques 4.2. Interaction with target cells

Several populations of EVs are released from cells and have over- EVs can interact and be internalized by cells thus constituting
lapping biophysical and biochemical properties. In view of this, the vehicles of biomolecule transfer and, consequently, provide the means
purification of homogeneous EVs populations constitutes a challenge at for communication among cells. In disease, they are involved in the
present time and several methodologies may be used [49,50]. The most spreading of toxic molecules from cell to cell underlying the transmis-
widely used approaches for EVs separation are based on differential sion of pathogenicity, which is relevant in cancer [1,2]. On the other
centrifugation, predominantly, ultracentrifugation of cell supernatants hand, EVs from healthy cells or from stem cells have beneficial effects
and biological fluids. For purity improvement and separation refine- on sick cells [62].
ment of different EVs populations, ultracentrifugation in gradients of EVs may modify target cell and organ function. For example, EVs
sucrose [8,51] or iodixanol [35] can be used. More recently, the interest from glioblastoma cells were shown to transmit mRNA, miRNA and
in size-exclusion chromatography for the separation of EVs has angiogenic proteins to brain microvascular endothelial cells in culture
increased [52]. In addition, affinity chromatography, including immu- [63]; microvesicles containing the oncogenic receptor EGFRvIII also
noisolation with antibodies against specific EVs components [35] allow transferred the oncogenic activity to cancer cells lacking EGFRvIII [64].
the recovery of more homogeneous populations based on EVs surface Moreover, EVs play different roles in tumorigenesis, extracellular
epitopes. In view of EVs enrichment in specific glycans, lectin affinity matrix remodeling, metastases, tumor angiogenesis and tumor immu-
chromatography is also used in their purification [49,53]. Particularly, nity [1,5]. Very importantly, EVs were found to promote the formation
it constitutes the basis for the diagnostic platform ELLSA™ (Enzyme of pre-metastatic niches in distant sites with the remodeling of
Linked Lectin Specific Assay) to isolate disease-specific exosomes from extracellular matrix in different cancers [65–67] (see Section 4.3.1).
body fluids. Heparin-affinity in combination with ultrafiltration can EVs are recognized by molecules at the surface of target cells and
also be used for the purification of EVs released from normal and cancer they can be internalized by various mechanisms including clathrin-
cells, and from human blood plasma [54]. mediated endocytosis, phagocytosis, macropinocytosis and plasma or
endosomal membrane fusion [42,68] (Fig. 2). Several proteins, in many
4. Glycoconjugates from extracellular vesicles cases glycosylated are relevant in EVs uptake, including, tetraspanins,
integrins and other cell adhesion molecules, proteoglycans and lectins
4.1. Biogenesis and sorting [68].
The HSPGs constitute cell-surface receptors for EVs endocytosis.
Syndecan HSPGs are involved in the biogenesis and cargo loading of HSPGs of the syndecan and glypican type were found to mediate the
exosomes via the intracellular adaptor syntenin and several regulators uptake of EVs by glioblastoma cells, in a process that was dependent on
that include Alix, the small GTPase ARF6, lipid-modifying enzyme the 2-O and N-sulfation groups of the HSPGs [48]; uptake was
PLD2 and heparanase [55]. Heparanase caused enhanced exosome specifically inhibited by free heparan sulfate chains, whereas closely
secretion and altered exosome composition and function in different related chondroitin sulfate had no effect. HSPGs were also relevant for
human cancer cells [56]. More recently, it was shown that heparanase the uptake of EVs by hepatic stellate cells [69]. Furthermore, heparin
stimulated the production of exosomes with syntenin-1, syndecan and partially inhibited EVs uptake by bladder cancer cells [70]. HSPGs from
CD63, but not CD9, CD81 or flotillin-1 [57]. The underlying mechanism EVs of myeloma cell lines or myeloma patients interacted with HSPGs
proposed that syndecan clustered upon heparanase and protease from tumor cells or marrow stromal cells via fibronectin [71].
cleavage, which resulted in enhanced formation of intraluminal vesicles Lectins are also involved in exosome uptake. For example, galectin-
(in multivesicular endosomes), subsequently released as exosomes [55]. 5 from exosomes of rat reticulocyte was suggested to mediate exosome
Other sorting mechanisms have been proposed based on the uptake by macrophages [60]. On the other hand, exosomes were taken
observations that EVs are strongly glycosylated and enriched in specific up by dendritic cells in a process involving the interaction between a C-
glycoproteins and glycans [6–8,16,42] (Section 3.1). Supporting this type lectin and mannose/glucosamine-rich C-type lectin receptor [72].
idea was the finding that complex N-glycosylation of the glycoprotein B-cell derived exosomes displayed sialic acid α2,3-linked on their
EWI-2 was essential for its sorting into EVs in a melanoma cell line surface that mediated the binding to sialoadhesin (CD169, Siglec1)
using the N-glycosylation inhibitor deoxymannojirymycin and by from macrophages in spleen and lymph node [73], therefore, playing a
mutating EWI-2 N-glycosylation sites [58]. Other studies reported that role in immune response.
kifunensin, an inhibitor of glycoprotein processing from high mannose Lamp2b fusion proteins displayed at the surface of exosomes have
to complex-type N-glycans, affected the composition of EVs from been used as a means to improve cell targeting. The N-glycosylation
ovarian carcinoma cells [16]. engineering of those surface proteins through the introduction of N-
Glycan-based sorting of glycoproteins into exosomes could involve glycosylation sites was shown to increase their stability having as
cellular lectins, such as galectins. Galectin-3 and galectin-4 were found consequence an enhanced uptake by recipient neuroblastoma cells
in EVs [7,59] and inhibition of glycosylation caused decreased levels of [74].

161
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

4.3. Cancer glyco-biomarkers other hand, glycomics of EVs glycoproteins from SKOV3 and OVMz
ovarian cancer cells showed the presence of bisecting GlcNAc-contain-
EVs transport cellular material to the extracellular environment, are ing-glycans and high mannose glycans [8,16]. These structures were
capable of crossing biological barriers and are found in biological fluids proposed to constitute ovarian cancer biomarkers, which was in
including blood, urine and cerebrospinal fluid. Therefore, they consti- agreement with the upregulation of bisecting GlcNAc in tissues from
tute valuable targets for biomarker identification. Potential biomarkers patients [27] and predominance of high mannose-type glycans in tumor
have been identified in EVs from glioblastoma, urogenital cancers tissues region [85].
including bladder and prostate cancer, melanoma, colorectal, breast, LGALS3BP is a heavily glycosylated protein with sialic acid and
ovarian, pancreatic cancers and lung adenocarcinoma [75–77]. complex N-glycans, that is associated with EVs from different cancer
Furthermore, the enhanced levels of EVs in body fluids from cancer cells [8,16,86] (EVPedia, Vesiclepedia, ExoCarta). In ovarian carcinoma
patients have been proposed as potential biomarkers [78]. cells it was found to contain α2,3-linked sialic acid based on lectin
EVs are highly glycosylated since they are derived from the plasma blotting with Maackia amurensis lectin and digestion with Streptococcus
membrane or from multivesicular endosomes. In cancer, EVs carry pneumoniae sialidase [8,16,42]. LGALS3BP is an extracellular matrix
molecules expressed by tumor cells, including glycoconjugates, to the protein and binds galectin-3 as well as other glycoproteins from the
extracellular environment and in this context EVs are considered as extracellular matrix including integrins; these interactions could pro-
surrogates of the originating tumor cells that are found in body fluids. vide the basis for its localization in EVs. LGALS3BP has been proposed
The proportions of the different molecules may be specific to EVs in as a potential cancer biomarker, for example, in pancreatic carcinoma
view of specific sorting events that take place during EVs biogenesis. [87] and prostate cancer [88].
For example, the enrichment of specific molecules, which are used as In breast cancer a highly glycosylated form of the extracellular
EVs markers (such as, glycosylated tetraspanin CD63, sialoglycoprotein matrix metalloproteinase inducer (EMMPRIN) was found as a marker
LGALS3BP) has been observed; furthermore, characteristic glycosigna- for proinvasive EVs from tumor cells. Furthermore, EMMPRIN was
tures of EVs relatively to total cellular membranes [7,16], or to plasma present at high levels on EVs from metastatic breast cancer patients in
membrane [8] also suggest glycan-based sorting to EVs possibly vivo. Anti-EMMPRIN strategies, such as EV de-N-deglycosylation with
involving intracellular lectins (Section 4.1). Considering that changes peptide N-glycosidase F, gene knockdown, and specific blocking
in glycosylation are a hallmark of cancer (Section 2.3), glycoconjugates peptides, inhibited EV-induced invasion [89]; therefore, EMMPRIN
from EVs and their respective glycosylation could constitute potential glycosylation appeared relevant for tumor cell phenotype.
biomarker targets in cancer. In this context two major aspects may be O-GlcNAcylation is a post-translational modification mainly of
considered: the identification of potential novel glyco-biomarkers and nuclear, cytoplasmic and mitochondrial proteins but recently it has
the improvement of glyco-biomarkers already used in the clinics. These also been found in proteins from EVs. Furthermore, O-GlcNAcylation of
issues will be discussed in the next two sections. many EV proteins from colorectal cancer cells was increased in
metastatic cells, thus suggesting its potential as biomarker [90]. In this
4.3.1. Glycoconjugates from extracellular vesicles as potential cancer study the proteins transitional endoplasmic reticulum ATPase and
biomarkers RuVB-like 1 contained O-GlcNAc and their levels were significantly
Recently, several studies have addressed the potential of EVs increased in EVs from metastatic cells.
glycoconjugates and their glycosylation as biomarkers in cancer A recent body of evidence has demonstrated that exosomes have the
(Table 1). For example, in glioblastoma EVs elevated expression of potential to form pre-metastatic niches creating a favorable environ-
several proteins including the N-glycosylated receptors EGFR and ment in target organs for metastases formation. Exosomes from highly
EGFRvIII were detected, and EVs were shown to constitute a surrogate metastatic melanoma increased the metastatic behavior of primary
of primary tumor mutations and to be useful in monitoring the effects of tumors and an exosome-specific melanoma signature with prognostic
treatment [79]. EGFR overexpression is detected in a high number of and therapeutic potential, comprised of TYRP2, VLA-4 (integrin α4β1),
glioblastoma multiforme cases, and EGFRvIII, a genomic deletion HSP70, an HSP90 isoform and the MET oncoprotein was found [65].
variant of EGFR, which is constitutively active and highly oncogenic More recently, in pancreatic cancer it was described that the formation
is also often expressed. Mutations in EGFR that result in deregulation of of the pre-metastatic niche involved upregulation of macrophage
its activity are also present in other types of cancer and several studies migration inhibitory factor (elevated in plasma exosomes from pan-
describing its glycosylation and association with cancer have been creatic cancer patients), which induced expression of the N-linked
performed, for example, glycomic analysis showed that EGFR-specific glycoprotein transforming growth factor beta (TGF-beta) by Kupffer
N-glycan signatures included high bisecting β1,4-GlcNAcylation and cells, which in turn, led to the production of the N- and O-linked
low α2,3-sialylation relative to EGFR-negative colorectal cancer tissues glycoprotein fibronectin by hepatic stellate cells [66]. In this context,
[80], but the relevance of EGFR glycosylation from EVs has been less integrins play a very relevant role; more specifically, tumor exosome
explored. integrins determined organotropic metastasis (integrins α6β4 and α6β1
In ovarian cancer patients, malignant ascites-derived exosomes associated with lung metastasis, integrin αvβ5 associated with liver
contain the N- and O-glycosylated GPI-anchor CD24 and the N- metastasis), and clinical data from patients with breast or pancreatic
glycosylated epithelial cell adhesion molecule EpCAM that are impor- cancer indicated that exosomal integrins could be used to predict organ-
tant for diagnostics [81,82]. CD24 is a well-established marker not only specific metastasis [67]. Integrins are N-glycosylated proteins that
in ovarian but also other types of cancer where it is associated with a exhibit glycosylation changes in cancer with implications in malig-
poor prognosis; it was also detected in some tumor stem cells [82]. Anti- nancy [24], and a particularly interesting topic would be to explore
CD24 antibody SWA11 studies using A549 lung and SKOV3 ovarian glycosylation of EVs integrins in depth.
carcinoma cells in mouse showed that CD24 targeting increased Glycolipids from EVs have also been suggested as potential cancer
infiltration of tumor tissues with immune cells suggesting involvement biomarkers elsewhere [47].
of antibody-dependent cell-mediated cytotoxicity and strong alterations Summing-up, there is a recent constellation of observations that
in intratumoral cytokine microenvironment; moreover, combination of point to EVs glycoconjugates/glycosignatures as emerging targets with
SWA11 mAb with gemcitabine treatment strongly potentiated its anti- potential in the cancer biomarker field.
cancer efficacy in A549 lung cancer model [83]. CD24 glycans from
SKOV3 cells contained α2,3/6-linked sialic acid and were recognized by 4.3.2. Detection of clinical glyco-biomarkers in extracellular vesicles
Siglec-5 and P-selectin [84], but further studies are required to Several glycoconjugates already used in the clinics as cancer
elucidate in detail its glycosignatures and relevance in cancer. On the biomarkers have been detected in EVs (Table 2). Selected glyco-

162
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

Table 2
Selected glycoconjugate/glycan tumor markers found in EVs.
Tumor markers were extracted from https://www.cancer.gov/about-cancer/diagnosis-staging/diagnosis/tumor-markers-fact-sheet. Type of glycosylation was extracted from http://
www.uniprot.org. The identification of the targets in the EVs databases was only considered from human origin and it was not necessarily associated with the type of cancer for which the
glycoconjugate is a biomarker. Date: 2nd February 2017.

Tumor markers used in the clinics Identification in EVs

Marker Cancer Biofluid/ Uniprot Glycosylation type EVPedia Vesiclepedia ExoCarta


tumor cells entry

Alpha-fetoprotein (AFP) Liver and germ cell tumors Blood P02771 N-glycosylated + VP_174 174
Beta-human chorionic gonadotropin Choriocarcinoma, germ cell Urine, blood P0DN86 N- and O-glycosylated − VP_1082 1082
(Beta-hCG) tumors
C-kit/CD117 Gastrointestinal stromal tumor, Tumor P10721 N-glycosylated − VP_3815 3815
mucosal melanoma
CA15-3/CA27.29 (MUC1) Breast Blood P15941 N- and O-glycosylated + VP_4582 4582
CA19-9 Pancreatic, gallbladder, bile Blood − Glycan sialyl-Lewisa [94]
duct, gastric
CA-125 (MUC16) Ovary Blood Q8WXI7 N- and O-glycosylated + VP_94,025 94025
Carcinoembryonic antigen (CEA) Colorectal and others Blood P06731 N-glycosylated + VP_1048 1048
Chromogranin A (CgA) Neuroendocrine tumors Blood P10645 O-glycosylated + VP_1113 1113
Estrogen receptor (ER) Breast Tumor P03372 O-GlcNAc + VP_2099 2099
HE4 Ovary Blood Q14508 N-glycosylated + VP_10406 10406
HER2/neu Breast, gastric, gastroesophageal Tumor P04626 N-glycosylated + VP_2064 2064
junction adenocarcinoma
Prostate-specific antigen (PSA) Prostate Blood P07288 N-glycosylated + VP_354 354
Urokinase plasminogen activator (uPA), Breast Tumor P00749, O-Fucose/N- +/+ VP_5328, VP- 5328/5054
plasminogen activator inhibitor (PAI- P05121 glycosylated, N- 5054
1) glycosylated

biomarkers that are membrane bound will be discussed in the next The human epidermal growth factor receptor 2 (HER2, also known
paragraphs. as ErBB2 or HER2/neu) is a type 1 transmembrane glycoprotein with an
CA15-3 and CA27.29 are epitopes from mucin 1 (MUC1) that are extracellular ectodomain and an intracellular domain with tyrosine
used in breast cancer diagnosis. MUC1 is a type I transmembrane kinase activity. HER2 is overexpressed in several types of cancer,
protein with a heavily O- and N-glycosylated extracellular domain. particularly, in breast cancer [101]. Recently, it was found that cancer
Aberrantly glycosylated MUC1 is overexpressed in most human epithe- cell-derived HER2 exosomes could stimulate the MAPK pathway in
lial cancers, including breast cancer [18]. MUC1 was found in lipid rafts monocytes with possible implications in the formation of tumor-
from plasma membrane and exosomes of human breast carcinoma cells associated macrophages [102]. Further data suggested that HER2-
MCF-7 [91]. positive exosomes could decrease the efficacy of antibody Trastuzumab
Carbohydrate antigen 19-9 (CA19-9) corresponds to the sialylated therapy [103].
fucosylated structure sialyl-Lewisa present on glycolipids and glycopro- These glyco-biomarkers used in clinical practice usually have
teins. CA19-9 is a widely used biomarker in pancreatic cancer, although sensitivity and specificity limitations when used alone. However, an
it presents limited sensitivity and specificity. It has been tested in interesting aspect would be their screening in EVs, which could
combination with other biomarkers; for example, CA19-9 and CA125 potentially improve their predictive value. Of particular interest would
have good sensitivity for detecting preclinical pancreatic cancer and also be to investigate if disease-associated glycoforms are enriched in
can be used to evaluate prognosis [92,93]. Recently, CA19-9 has been EVs from body fluids.
detected in pancreatic cancer derived exosomes [94].
CA-125 is an ovarian cancer antigen that is screened in the serum of
patients as a biomarker. It is present in mucin 16 (MUC16), which is a 5. Conclusions and future perspectives
highly N- and O-glycosylated type I membrane glycoprotein with a
large extracellular domain. This biomarker alone has specificity limita- The study of EVs has been subject of increased interest in recent
tions but it is useful for monitoring the effectiveness of treatment and years and the recognized role of EVs in transmitting biomolecules
detecting recurrence [95]. More recently, approaches involving the among cells as well as their potential to provide disease biomarkers are
glycoprofiling of MUC16 have shown promising results towards in- particularly relevant. Several aspects currently remain a challenge, such
creased biomarker sensitivity for ovarian cancer [96]. Furthermore, the as purification of homogeneous populations of vesicles, regulation of
screening for CA-125, EpCAM and CD24 using a microfluidic approach EVs release and modulation of recognition and uptake of EVs by target
on the ExoSearch chip, which provides an enriched preparation of cells.
blood plasma exosomes, showed significant diagnostic power for Glycoconjugates and their glycosylation in EVs could emerge as
ovarian cancer [97]. useful tools for major advances in the field. For example, a useful
Carcinoembryonic antigen (CEA) belongs to the immunoglobulin approach for EVs fractionation in view of their diversity, is to explore
superfamily, it is heavily N-glycosylated and is bound to the plasma their characteristic glycosylation properties as the basis for affinity
membrane via a GPI-anchor. CEA is the main marker in colorectal purification. Concerning biogenesis, the HSPG syndecan participates in
cancer, in prognosis and postoperative surveillance and for therapy exosome formation and cargo loading. Furthermore, characteristic
monitoring [98]. CEA has been detected in EVs from colorectal cancer glycosignatures from EVs suggest that novel specific mechanisms,
cells and from plasma of patients with colorectal cancer [99]. Recently, possibly related with intracellular lectins, participate in glycoprotein
exosomes from colorectal cancer cells, which were CEA positive, were sorting into EVs.
found to induce morphological and functional changes in colonic In the context of EVs interaction with other cells, HSPGs from host
mesenchymal stromal cells, possibly favoring tumor growth and cells play a well-recognized role, and the binding partners from EVs to
malignant progression [100]. HSPGs are currently an interesting topic of research. EVs have been
largely explored as therapeutic vehicles of delivery (e.g., drugs, nucleic

163
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

acids, proteins) in cancer and in neurodegenerative diseases. Glycobiology 21 (2011) 376–386.


[16] J. Gomes, P. Gomes-Alves, S.B. Carvalho, C. Peixoto, P.M. Alves, P. Altevogt,
Modulation of EVs surface glycosylation could be a promising strategy J. Costa, Extracellular vesicles from ovarian carcinoma cells display specific
for protein stabilization and to improve interactions with target cells. glycosignatures, Biomol. Ther. 5 (2015) 1741–1761.
EVs also hold a great potential for the identification of disease [17] Y.H. Ahn, J.Y. Kim, J.S. Yoo, Quantitative mass spectrometric analysis of
glycoproteins combined with enrichment methods, Mass Spectrom. Rev. 34 (2015)
biomarkers since stressed cells release EVs that are found in body fluids, 148–165.
such as plasma, urine or cerebrospinal fluid; therefore, EVs are [18] S.R. Stowell, T. Ju, R.D. Cummings, Protein glycosylation in cancer, Annu. Rev.
considered surrogates of the originating cells. In cancer major altera- Pathol. 10 (2015) 473–510.
[19] J. Costa, E. Grabenhorst, M. Nimtz, H.S. Conradt, Stable expression of the Golgi
tions in cell surface glycosylation take place, which will be reflected at form and secretory variants of human fucosyltransferase III from BHK-21 cells.
the surface of EVs. Therefore, the study of glycoconjugates and their Purification and characterization of an engineered truncated form from the culture
glycosylation from body fluids EVs constitutes a promising strategy to medium, J. Biol. Chem. 272 (1997) 11613–11621.
[20] M.R. Kudelka, A. Antonopoulos, Y. Wang, D.M. Duong, X. Song, N.T. Seyfried,
identify novel cancer markers. In addition, many cancer markers
A. Dell, S.M. Haslam, R.D. Cummings, T. Ju, Cellular O-glycome reporter/
already used in the clinics are glycosylated and also found in EVs; the amplification to explore O-glycans of living cells, Nat. Methods 13 (2016) 81–86.
investigation of these clinical biomarkers specifically in EVs may [21] C.J. Gray, B. Thomas, R. Upton, L.G. Migas, C.E. Eyers, P.E. Barran, S.L. Flitsch,
improve their predictive value. Applications of ion mobility mass spectrometry for high throughput, high
resolution glycan analysis, Biochim. Biophys. Acta 1860 (2016) 1688–1709.
In conclusion, glycosylation appears to be highly relevant in the [22] L.Y. Lee, M. Thaysen-Andersen, M.S. Baker, N.H. Packer, W.S. Hancock,
extracellular vesicle field. Particularly in cancer it holds great promise S. Fanayan, Comprehensive N-glycome profiling of cultured human epithelial
towards novel biomarker identification as well as improvement of breast cells identifies unique secretome N-glycosylation signatures enabling
tumorigenic subtype classification, J. Proteome Res. 13 (2014) 4783–4795.
clinical biomarkers. [23] A. Kobata, Exo- and endoglycosidases revisited, Proc. Jpn. Acad. Ser. B Phys. Biol.
Sci. 89 (2013) 97–117.
Transparency Document [24] S.S. Pinho, C.A. Reis, Glycosylation in cancer: mechanisms and clinical implica-
tions, Nat. Rev. Cancer 15 (2015) 540–555.
[25] M.J. Kailemia, D. Park, C.B. Lebrilla, Glycans and glycoproteins as specific
The http://dx.doi.org/10.1016/j.bbcan.2017.03.007 associated biomarkers for cancer, Anal. Bioanal. Chem. (2016).
with this article can be found in the online version. [26] N. Taniguchi, Y. Kizuka, Glycans and cancer: role of N-glycans in cancer
biomarker, progression and metastasis, and therapeutics, Adv. Cancer Res. 126
(2015) 11–51.
Acknowledgments [27] H. Allam, K. Aoki, B.B. Benigno, J.F. McDonald, S.G. Mackintosh, M. Tiemeyer,
K.L. Abbott, Glycomic analysis of membrane glycoproteins with bisecting glyco-
sylation from ovarian cancer tissues reveals novel structures and functions, J.
We thank Prof. Peter Altevogt, DKFZ, Germany, for critical reading
Proteome Res. 14 (2015) 434–446.
of the manuscript and useful suggestions. This work was supported by [28] M. Anugraham, F. Jacob, S. Nixdorf, A.V. Everest-Dass, V. Heinzelmann-Schwarz,
Euronanomed 2 ERA-NET, project GlioEx, Fundação para a Ciência e a N.H. Packer, Specific glycosylation of membrane proteins in epithelial ovarian
Tecnologia (FCT), Portugal, grant ENMed/0001/2013; FCT/Ministério cancer cell lines: glycan structures reflect gene expression and DNA methylation
status, Mol. Cell. Proteomics 13 (2014) 2213–2232.
da Educação e Ciência, grant iNOVA4Health-UID/Multi/04462/2013. [29] T. Nakagawa, E. Miyoshi, T. Yakushijin, N. Hiramatsu, T. Igura, N. Hayashi,
N. Taniguchi, A. Kondo, Glycomic analysis of alpha-fetoprotein L3 in hepatoma
References cell lines and hepatocellular carcinoma patients, J. Proteome Res. 7 (2008)
2222–2233.
[30] A. Harduin-Lepers, M.A. Krzewinski-Recchi, F. Colomb, F. Foulquier, S. Groux-
[1] M. Tkach, C. Thery, Communication by extracellular vesicles: where we are and Degroote, P. Delannoy, Sialyltransferases functions in cancers, Front. Biosci. (Elite
where we need to go, Cell 164 (2016) 1226–1232. Ed.) 4 (2012) 499–515.
[2] A. Becker, B.K. Thakur, J.M. Weiss, H.S. Kim, H. Peinado, D. Lyden, Extracellular [31] A.S. Carvalho, A. Harduin-Lepers, A. Magalhaes, E. Machado, N. Mendes,
vesicles in cancer: cell-to-cell mediators of metastasis, Cancer Cell 30 (2016) L.T. Costa, R. Matthiesen, R. Almeida, J. Costa, C.A. Reis, Differential expression of
836–848. alpha-2,3-sialyltransferases and alpha-1,3/4-fucosyltransferases regulates the le-
[3] M. Nawaz, F. Fatima, K.C. Vallabhaneni, P. Penfornis, H. Valadi, K. Ekstrom, vels of sialyl Lewis a and sialyl Lewis x in gastrointestinal carcinoma cells, Int. J.
S. Kholia, J.D. Whitt, J.D. Fernandes, R. Pochampally, J.A. Squire, G. Camussi, Biochem. Cell Biol. 42 (2010) 80–89.
Extracellular vesicles: evolving factors in stem cell biology, Stem Cells Int. 2016 [32] J.R. Couchman, H. Multhaupt, R.D. Sanderson, Recent insights into cell surface
(2016) 1073140. heparan sulphate proteoglycans and cancer, F1000Res 5 (2016).
[4] L. Alvarez-Erviti, Y. Seow, H. Yin, C. Betts, S. Lakhal, M.J. Wood, Delivery of [33] M. Ahmed, N.K. Cheung, Engineering anti-GD2 monoclonal antibodies for cancer
siRNA to the mouse brain by systemic injection of targeted exosomes, Nat. immunotherapy, FEBS Lett. 588 (2014) 288–297.
Biotechnol. 29 (2011) 341–345. [34] M. Colombo, G. Raposo, C. Thery, Biogenesis, secretion, and intercellular
[5] R. Kalluri, The biology and function of exosomes in cancer, J. Clin. Invest. 126 interactions of exosomes and other extracellular vesicles, Annu. Rev. Cell Dev.
(2016) 1208–1215. Biol. 30 (2014) 255–289.
[6] L. Krishnamoorthy, J.W. Bess Jr., A.B. Preston, K. Nagashima, L.K. Mahal, HIV-1 [35] J. Kowal, G. Arras, M. Colombo, M. Jouve, J.P. Morath, B. Primdal-Bengtson,
and microvesicles from T cells share a common glycome, arguing for a common F. Dingli, D. Loew, M. Tkach, C. Thery, Proteomic comparison defines novel
origin, Nat. Chem. Biol. 5 (2009) 244–250. markers to characterize heterogeneous populations of extracellular vesicle sub-
[7] B.S. Batista, W.S. Eng, K.T. Pilobello, K.D. Hendricks-Munoz, L.K. Mahal, types, Proc. Natl. Acad. Sci. U. S. A. 113 (2016) E968–E977.
Identification of a conserved glycan signature for microvesicles, J. Proteome Res. [36] D.K. Kim, B. Kang, O.Y. Kim, D.S. Choi, J. Lee, S.R. Kim, G. Go, Y.J. Yoon,
10 (2011) 4624–4633. J.H. Kim, S.C. Jang, K.S. Park, E.J. Choi, K.P. Kim, D.M. Desiderio, Y.K. Kim,
[8] C. Escrevente, N. Grammel, S. Kandzia, J. Zeiser, E.M. Tranfield, H.S. Conradt, J. Lotvall, D. Hwang, Y.S. Gho, EVpedia: an integrated database of high-
J. Costa, Sialoglycoproteins and N-glycans from secreted exosomes of ovarian throughput data for systemic analyses of extracellular vesicles, J. Extracell.
carcinoma cells, PLoS One 8 (2013) e78631. Vesicles 2 (2013).
[9] J.Q. Gerlach, M.D. Griffin, Getting to know the extracellular vesicle glycome, Mol. [37] H. Kalra, R.J. Simpson, H. Ji, E. Aikawa, P. Altevogt, P. Askenase, V.C. Bond,
BioSyst. 12 (2016) 1071–1081. F.E. Borras, X. Breakefield, V. Budnik, E. Buzas, G. Camussi, A. Clayton,
[10] M.M. Fuster, J.D. Esko, The sweet and sour of cancer: glycans as novel therapeutic E. Cocucci, J.M. Falcon-Perez, S. Gabrielsson, Y.S. Gho, D. Gupta, H.C. Harsha,
targets, Nat. Rev. Cancer 5 (2005) 526–542. A. Hendrix, A.F. Hill, J.M. Inal, G. Jenster, E.M. Kramer-Albers, S.K. Lim,
[11] G. D'Angelo, S. Capasso, L. Sticco, D. Russo, Glycosphingolipids: synthesis and A. Llorente, J. Lotvall, A. Marcilla, L. Mincheva-Nilsson, I. Nazarenko,
functions, FEBS J. 280 (2013) 6338–6353. R. Nieuwland, E.N. Nolte-'t Hoen, A. Pandey, T. Patel, M.G. Piper, S. Pluchino,
[12] K. Marino, J. Bones, J.J. Kattla, P.M. Rudd, A systematic approach to protein T.S. Prasad, L. Rajendran, G. Raposo, M. Record, G.E. Reid, F. Sanchez-Madrid,
glycosylation analysis: a path through the maze, Nat. Chem. Biol. 6 (2010) R.M. Schiffelers, P. Siljander, A. Stensballe, W. Stoorvogel, D. Taylor, C. Thery,
713–723. H. Valadi, B.W. van Balkom, J. Vazquez, M. Vidal, M.H. Wauben, M. Yanez-Mo,
[13] M. Thaysen-Andersen, N.H. Packer, B.L. Schulz, Maturing glycoproteomics M. Zoeller, S. Mathivanan, Vesiclepedia: a compendium for extracellular vesicles
technologies provide unique structural insights into the N-glycoproteome and its with continuous community annotation, PLoS Biol. 10 (2012) e1001450.
regulation in health and disease, Mol. Cell. Proteomics 15 (2016) 1773–1790. [38] R.J. Simpson, H. Kalra, S. Mathivanan, ExoCarta as a resource for exosomal
[14] M.L. Silva, Cancer serum biomarkers based on aberrant post-translational research, J. Extracell. Vesicles 1 (2012).
modifications of glycoproteins: clinical value and discovery strategies, Biochim. [39] Z. Andreu, M. Yanez-Mo, Tetraspanins in extracellular vesicle formation and
Biophys. Acta 1856 (2015) 165–177. function, Front. Immunol. 5 (2014) 442.
[15] E. Machado, S. Kandzia, R. Carilho, P. Altevogt, H.S. Conradt, J. Costa, N- [40] D. Perez-Hernandez, C. Gutierrez-Vazquez, I. Jorge, S. Lopez-Martin, A. Ursa,
glycosylation of total cellular glycoproteins from the human ovarian carcinoma F. Sanchez-Madrid, J. Vazquez, M. Yanez-Mo, The intracellular interactome of
SKOV3 cell line and of recombinantly expressed human erythropoietin, tetraspanin-enriched microdomains reveals their function as sorting machineries

164
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

toward exosomes, J. Biol. Chem. 288 (2013) 11649–11661. V.R. Martins, J. Skog, R.N. Kaplan, M.S. Brady, J.D. Wolchok, P.B. Chapman,
[41] L.J. Vella, R.A. Sharples, V.A. Lawson, C.L. Masters, R. Cappai, A.F. Hill, Packaging Y. Kang, J. Bromberg, D. Lyden, Melanoma exosomes educate bone marrow
of prions into exosomes is associated with a novel pathway of PrP processing, J. progenitor cells toward a pro-metastatic phenotype through MET, Nat. Med. 18
Pathol. 211 (2007) 582–590. (2012) 883–891.
[42] C. Escrevente, S. Keller, P. Altevogt, J. Costa, Interaction and uptake of exosomes [66] B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, A. Becker,
by ovarian cancer cells, BMC Cancer 11 (2011) 108. A. Hoshino, M.T. Mark, H. Molina, J. Xiang, T. Zhang, T.M. Theilen, G. Garcia-
[43] M. Saraswat, S. Joenvaara, L. Musante, H. Peltoniemi, H. Holthofer, R. Renkonen, Santos, C. Williams, Y. Ararso, Y. Huang, G. Rodrigues, T.L. Shen, K.J. Labori,
N-linked (N-) glycoproteomics of urinary exosomes. [corrected], Mol. Cell. I.M. Lothe, E.H. Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. Grandgenett,
Proteomics 14 (2015) 263–276. M.A. Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, R.E. Schwartz,
[44] S. Staubach, P. Schadewaldt, U. Wendel, K. Nohroudi, F.G. Hanisch, Differential I. Matei, H. Peinado, B.Z. Stanger, J. Bromberg, D. Lyden, Pancreatic cancer
glycomics of epithelial membrane glycoproteins from urinary exovesicles reveals exosomes initiate pre-metastatic niche formation in the liver, Nat. Cell Biol. 17
shifts toward complex-type N-glycosylation in classical galactosemia, J. Proteome (2015) 816–826.
Res. 11 (2012) 906–916. [67] A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic Mark,
[45] J.Q. Gerlach, A. Kruger, S. Gallogly, S.A. Hanley, M.C. Hogan, C.J. Ward, L. Joshi, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop,
M.D. Griffin, Surface glycosylation profiles of urine extracellular vesicles, PLoS K. Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, T. Zhang,
One 8 (2013) e74801. H. Zhang, J. Hernandez, J.M. Weiss, V.D. Dumont-Cole, K. Kramer, L.H. Wexler,
[46] J. Echevarria, F. Royo, R. Pazos, L. Salazar, J.M. Falcon-Perez, N.C. Reichardt, A. Narendran, G.K. Schwartz, J.H. Healey, P. Sandstrom, K.J. Labori, E.H. Kure,
Microarray-based identification of lectins for the purification of human urinary P.M. Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur, M. Jain, K. Mallya,
extracellular vesicles directly from urine samples, Chembiochem 15 (2014) S.K. Batra, W.R. Jarnagin, M.S. Brady, O. Fodstad, V. Muller, K. Pantel, A.J. Minn,
1621–1626. M.J. Bissell, B.A. Garcia, Y. Kang, V.K. Rajasekhar, C.M. Ghajar, I. Matei,
[47] A. Llorente, T. Skotland, T. Sylvanne, D. Kauhanen, T. Rog, A. Orlowski, H. Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins determine
I. Vattulainen, K. Ekroos, K. Sandvig, Molecular lipidomics of exosomes released organotropic metastasis, Nature 527 (2015) 329–335.
by PC-3 prostate cancer cells, Biochim. Biophys. Acta 1831 (2013) 1302–1309. [68] L.A. Mulcahy, R.C. Pink, D.R. Carter, Routes and mechanisms of extracellular
[48] H.C. Christianson, K.J. Svensson, T.H. van Kuppevelt, J.P. Li, M. Belting, Cancer vesicle uptake, J. Extracell. Vesicles 3 (2014).
cell exosomes depend on cell-surface heparan sulfate proteoglycans for their [69] L. Chen, D.R. Brigstock, Integrins and heparan sulfate proteoglycans on hepatic
internalization and functional activity, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) stellate cells (HSC) are novel receptors for HSC-derived exosomes, FEBS Lett. 590
17380–17385. (2016) 4263–4274.
[49] E. Zeringer, T. Barta, M. Li, A.V. Vlassov, Strategies for isolation of exosomes, Cold [70] C.A. Franzen, P.E. Simms, A.F. Van Huis, K.E. Foreman, P.C. Kuo, G.N. Gupta,
Spring Harb. Protoc. 2015 (2015) 319–323. Characterization of uptake and internalization of exosomes by bladder cancer
[50] G. Pocsfalvi, C. Stanly, I. Fiume, K. Vekey, Chromatography and its hyphenation to cells, Biomed. Res. Int. 2014 (2014) 619829.
mass spectrometry for extracellular vesicle analysis, J. Chromatogr. A 1439 (2016) [71] A. Purushothaman, S.K. Bandari, J. Liu, J.A. Mobley, E.E. Brown, R.D. Sanderson,
26–41. Fibronectin on the surface of myeloma cell-derived exosomes mediates exosome-
[51] C. Escrevente, V.A. Morais, S. Keller, C.M. Soares, P. Altevogt, J. Costa, Functional cell interactions, J. Biol. Chem. 291 (2016) 1652–1663.
role of N-glycosylation from ADAM10 in processing, localization and activity of [72] S. Hao, O. Bai, F. Li, J. Yuan, S. Laferte, J. Xiang, Mature dendritic cells pulsed
the enzyme, Biochim. Biophys. Acta 1780 (2008) 905–913. with exosomes stimulate efficient cytotoxic T-lymphocyte responses and anti-
[52] C.S. Hong, S. Funk, L. Muller, M. Boyiadzis, T.L. Whiteside, Isolation of tumour immunity, Immunology 120 (2007) 90–102.
biologically active and morphologically intact exosomes from plasma of patients [73] S.C. Saunderson, A.C. Dunn, P.R. Crocker, A.D. McLellan, CD169 mediates the
with cancer, J. Extracell. Vesicles 5 (2016) 29289. capture of exosomes in spleen and lymph node, Blood 123 (2014) 208–216.
[53] F. Royo, P. Zuniga-Garcia, P. Sanchez-Mosquera, A. Egia, A. Perez, A. Loizaga, [74] M.E. Hung, J.N. Leonard, Stabilization of exosome-targeting peptides via engi-
R. Arceo, I. Lacasa, A. Rabade, E. Arrieta, R. Bilbao, M. Unda, A. Carracedo, neered glycosylation, J. Biol. Chem. 290 (2015) 8166–8172.
J.M. Falcon-Perez, Different EV enrichment methods suitable for clinical settings [75] T. Katsuda, N. Kosaka, T. Ochiya, The roles of extracellular vesicles in cancer
yield different subpopulations of urinary extracellular vesicles from human biology: toward the development of novel cancer biomarkers, Proteomics 14
samples, J. Extracell. Vesicles 5 (2016) 29497. (2014) 412–425.
[54] L. Balaj, N.A. Atai, W. Chen, D. Mu, B.A. Tannous, X.O. Breakefield, J. Skog, [76] S. Boukouris, S. Mathivanan, Exosomes in bodily fluids are a highly stable resource
C.A. Maguire, Heparin affinity purification of extracellular vesicles, Sci. Rep. 5 of disease biomarkers, Proteomics Clin. Appl. 9 (2015) 358–367.
(2015) 10266. [77] M. Nawaz, G. Camussi, H. Valadi, I. Nazarenko, K. Ekstrom, X. Wang, S. Principe,
[55] V. Friand, G. David, P. Zimmermann, Syntenin and syndecan in the biogenesis of N. Shah, N.M. Ashraf, F. Fatima, L. Neder, T. Kislinger, The emerging role of
exosomes, Biol. Cell. 107 (2015) 331–341. extracellular vesicles as biomarkers for urogenital cancers, Nat. Rev. Urol. 11
[56] C.A. Thompson, A. Purushothaman, V.C. Ramani, I. Vlodavsky, R.D. Sanderson, (2014) 688–701.
Heparanase regulates secretion, composition, and function of tumor cell-derived [78] F. Cappello, M. Logozzi, C. Campanella, C.C. Bavisotto, A. Marcilla, F. Properzi,
exosomes, J. Biol. Chem. 288 (2013) 10093–10099. S. Fais, Exosome levels in human body fluids: a tumor marker by themselves? Eur.
[57] B. Roucourt, S. Meeussen, J. Bao, P. Zimmermann, G. David, Heparanase activates J. Pharm. Sci. 96 (2017) 93–98.
the syndecan-syntenin-ALIX exosome pathway, Cell Res. 25 (2015) 412–428. [79] H. Shao, J. Chung, L. Balaj, A. Charest, D.D. Bigner, B.S. Carter, F.H. Hochberg,
[58] Y. Liang, W.S. Eng, D.R. Colquhoun, R.R. Dinglasan, D.R. Graham, L.K. Mahal, X.O. Breakefield, R. Weissleder, H. Lee, Protein typing of circulating microvesicles
Complex N-linked glycans serve as a determinant for exosome/microvesicle cargo allows real-time monitoring of glioblastoma therapy, Nat. Med. 18 (2012)
recruitment, J. Biol. Chem. 289 (2014) 32526–32537. 1835–1840.
[59] J.L. Welton, S. Khanna, P.J. Giles, P. Brennan, I.A. Brewis, J. Staffurth, [80] M.K. Sethi, H. Kim, C.K. Park, M.S. Baker, Y.K. Paik, N.H. Packer, W.S. Hancock,
M.D. Mason, A. Clayton, Proteomics analysis of bladder cancer exosomes, Mol. S. Fanayan, M. Thaysen-Andersen, In-depth N-glycome profiling of paired color-
Cell. Proteomics 9 (2010) 1324–1338. ectal cancer and non-tumorigenic tissues reveals cancer-, stage- and EGFR-specific
[60] C. Barres, L. Blanc, P. Bette-Bobillo, S. Andre, R. Mamoun, H.J. Gabius, M. Vidal, protein N-glycosylation, Glycobiology 25 (2015) 1064–1078.
Galectin-5 is bound onto the surface of rat reticulocyte exosomes and modulates [81] H. Im, H. Shao, Y.I. Park, V.M. Peterson, C.M. Castro, R. Weissleder, H. Lee, Label-
vesicle uptake by macrophages, Blood 115 (2010) 696–705. free detection and molecular profiling of exosomes with a nano-plasmonic sensor,
[61] E. Honig, K. Schneider, R. Jacob, Recycling of galectin-3 in epithelial cells, Eur. J. Nat. Biotechnol. 32 (2014) 490–495.
Cell Biol. 94 (2015) 309–315. [82] S. Runz, S. Keller, C. Rupp, A. Stoeck, Y. Issa, D. Koensgen, A. Mustea, J. Sehouli,
[62] T. Lener, M. Gimona, L. Aigner, V. Borger, E. Buzas, G. Camussi, N. Chaput, G. Kristiansen, P. Altevogt, Malignant ascites-derived exosomes of ovarian
D. Chatterjee, F.A. Court, H.A. Del Portillo, L. O'Driscoll, S. Fais, J.M. Falcon- carcinoma patients contain CD24 and EpCAM, Gynecol. Oncol. 107 (2007)
Perez, U. Felderhoff-Mueser, L. Fraile, Y.S. Gho, A. Gorgens, R.C. Gupta, 563–571.
A. Hendrix, D.M. Hermann, A.F. Hill, F. Hochberg, P.A. Horn, D. de Kleijn, [83] A.V. Salnikov, N.P. Bretz, C. Perne, J. Hazin, S. Keller, M. Fogel, I. Herr,
L. Kordelas, B.W. Kramer, E.M. Kramer-Albers, S. Laner-Plamberger, S. Laitinen, T. Schlange, G. Moldenhauer, P. Altevogt, Antibody targeting of CD24 efficiently
T. Leonardi, M.J. Lorenowicz, S.K. Lim, J. Lotvall, C.A. Maguire, A. Marcilla, retards growth and influences cytokine milieu in experimental carcinomas, Br. J.
I. Nazarenko, T. Ochiya, T. Patel, S. Pedersen, G. Pocsfalvi, S. Pluchino, Cancer 108 (2013) 1449–1459.
P. Quesenberry, I.G. Reischl, F.J. Rivera, R. Sanzenbacher, K. Schallmoser, [84] G. Kristiansen, E. Machado, N. Bretz, C. Rupp, K.J. Winzer, A.K. Konig,
I. Slaper-Cortenbach, D. Strunk, T. Tonn, P. Vader, B.W. van Balkom, M. Wauben, G. Moldenhauer, F. Marme, J. Costa, P. Altevogt, Molecular and clinical dissection
S.E. Andaloussi, C. Thery, E. Rohde, B. Giebel, Applying extracellular vesicles of CD24 antibody specificity by a comprehensive comparative analysis, Lab.
based therapeutics in clinical trials - an ISEV position paper, J. Extracell. Vesicles 4 Investig. 90 (2010) 1102–1116.
(2015) 30087. [85] A.V. Everest-Dass, M.T. Briggs, G. Kaur, M.K. Oehler, P. Hoffmann, N.H. Packer, N-
[63] J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves, glycan MALDI imaging mass spectrometry on formalin-fixed paraffin-embedded
W.T. Curry Jr., B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma tissue enables the delineation of ovarian cancer tissues, Mol. Cell. Proteomics 15
microvesicles transport RNA and proteins that promote tumour growth and (2016) 3003–3016.
provide diagnostic biomarkers, Nat. Cell Biol. 10 (2008) 1470–1476. [86] S.N. Hurwitz, M.A. Rider, J.L. Bundy, X. Liu, R.K. Singh, D.G. Meckes Jr.,
[64] K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A. Guha, J. Rak, Proteomic profiling of NCI-60 extracellular vesicles uncovers common protein
Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived cargo and cancer type-specific biomarkers, Oncotarget (2016).
from tumour cells, Nat. Cell Biol. 10 (2008) 619–624. [87] B.M. Kunzli, P.O. Berberat, Z.W. Zhu, M. Martignoni, J. Kleeff, A.A. Tempia-
[65] H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, G. Moreno- Caliera, M. Fukuda, A. Zimmermann, H. Friess, M.W. Buchler, Influences of the
Bueno, M. Hergueta-Redondo, C. Williams, G. Garcia-Santos, C. Ghajar, lysosomal associated membrane proteins (lamp-1, lamp-2) and mac-2 binding
A. Nitadori-Hoshino, C. Hoffman, K. Badal, B.A. Garcia, M.K. Callahan, J. Yuan, protein (mac-2-BP) on the prognosis of pancreatic carcinoma, Cancer 94 (2002)

165
J. Costa BBA - Reviews on Cancer 1868 (2017) 157–166

228–239. U. Mandel, H. Clausen, A. Felix, N. Lunet, L. David, Detection of glyco-mucin


[88] J. Hu, J. He, Y. Kuang, Z. Wang, Z. Sun, H. Zhu, X. Liu, Expression and significance profiles improves specificity of MUC16 and MUC1 biomarkers in ovarian serous
of 90K/Mac-2BP in prostate cancer, Exp. Ther. Med. 5 (2013) 181–184. tumours, Mol. Oncol. 9 (2015) 503–512.
[89] K. Menck, C. Scharf, A. Bleckmann, L. Dyck, U. Rost, D. Wenzel, V.M. Dhople, [97] Z. Zhao, Y. Yang, Y. Zeng, M. He, A microfluidic ExoSearch chip for multiplexed
L. Siam, T. Pukrop, C. Binder, F. Klemm, Tumor-derived microvesicles mediate exosome detection towards blood-based ovarian cancer diagnosis, Lab Chip 16
human breast cancer invasion through differentially glycosylated EMMPRIN, J. (2016) 489–496.
Mol. Cell Biol. 7 (2015) 143–153. [98] M.J. Duffy, R. Lamerz, C. Haglund, A. Nicolini, M. Kalousova, L. Holubec,
[90] P. Chaiyawat, C. Weeraphan, P. Netsirisawan, D. Chokchaichamnankit, C. Sturgeon, Tumor markers in colorectal cancer, gastric cancer and gastroin-
C. Srisomsap, J. Svasti, V. Champattanachai, Elevated O-GlcNAcylation of extra- testinal stromal cancers: European group on tumor markers 2014 guidelines
cellular vesicle proteins derived from metastatic colorectal cancer cells, Cancer update, Int. J. Cancer 134 (2014) 2513–2522.
Genomics Proteomics 13 (2016) 387–398. [99] V. Huber, S. Fais, M. Iero, L. Lugini, P. Canese, P. Squarcina, A. Zaccheddu,
[91] S. Staubach, H. Razawi, F.G. Hanisch, Proteomics of MUC1-containing lipid rafts M. Colone, G. Arancia, M. Gentile, E. Seregni, R. Valenti, G. Ballabio, F. Belli,
from plasma membranes and exosomes of human breast carcinoma cells MCF-7, E. Leo, G. Parmiani, L. Rivoltini, Human colorectal cancer cells induce T-cell death
Proteomics 9 (2009) 2820–2835. through release of proapoptotic microvesicles: role in immune escape,
[92] T. Kamisawa, L.D. Wood, T. Itoi, K. Takaori, Pancreatic cancer, Lancet 388 (2016) Gastroenterology 128 (2005) 1796–1804.
73–85. [100] L. Lugini, M. Valtieri, C. Federici, S. Cecchetti, S. Meschini, M. Condello,
[93] D.P. O'Brien, N.S. Sandanayake, C. Jenkinson, A. Gentry-Maharaj, S. Apostolidou, M. Signore, S. Fais, Exosomes from human colorectal cancer induce a tumor-like
E.O. Fourkala, S. Camuzeaux, O. Blyuss, R. Gunu, A. Dawnay, A. Zaikin, behavior in colonic mesenchymal stromal cells, Oncotarget 7 (2016)
R.C. Smith, I.J. Jacobs, U. Menon, E. Costello, S.P. Pereira, J.F. Timms, Serum 50086–50098.
CA19-9 is significantly upregulated up to 2 years before diagnosis with pancreatic [101] F. Montemurro, M. Scaltriti, Biomarkers of drugs targeting HER-family signalling
cancer: implications for early disease detection, Clin. Cancer Res. 21 (2015) in cancer, J. Pathol. 232 (2014) 219–229.
622–631. [102] X. Song, Y. Ding, G. Liu, X. Yang, R. Zhao, Y. Zhang, X. Zhao, G.J. Anderson,
[94] N. Javeed, G. Sagar, S.K. Dutta, T.C. Smyrk, J.S. Lau, S. Bhattacharya, M. Truty, G. Nie, Cancer cell-derived exosomes induce mitogen-activated protein kinase-
G.M. Petersen, R.J. Kaufman, S.T. Chari, D. Mukhopadhyay, Pancreatic cancer- dependent monocyte survival by transport of functional receptor tyrosine kinases,
derived exosomes cause paraneoplastic beta-cell dysfunction, Clin. Cancer Res. 21 J. Biol. Chem. 291 (2016) 8453–8464.
(2015) 1722–1733. [103] V. Ciravolo, V. Huber, G.C. Ghedini, E. Venturelli, F. Bianchi, M. Campiglio,
[95] Z. Su, W.S. Graybill, Y. Zhu, Detection and monitoring of ovarian cancer, Clin. D. Morelli, A. Villa, P. Della Mina, S. Menard, P. Filipazzi, L. Rivoltini,
Chim. Acta 415 (2013) 341–345. E. Tagliabue, S.M. Pupa, Potential role of HER2-overexpressing exosomes in
[96] S. Ricardo, L. Marcos-Silva, D. Pereira, R. Pinto, R. Almeida, O. Soderberg, countering trastuzumab-based therapy, J. Cell. Physiol. 227 (2012) 658–667.

166

You might also like