You are on page 1of 12

G Model

YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS


Seminars in Cancer Biology xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


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

Review

The role of exosomes in cancer metastasis


Teresa Bernadette Steinbichler a , József Dudás a , Herbert Riechelmann a ,
Ira-Ida Skvortsova b,∗
a
Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
b
Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria

a r t i c l e i n f o a b s t r a c t

Article history: Exosomes are small membrane vesicles with a size ranging from 40 to 100 nm. They can serve as functional
Received 20 December 2016 mediators in cell interaction leading to cancer metastasis. Metastasis is a complex multistep process of
Accepted 9 February 2017 cancer cell invasion, survival in blood vessels, attachment to and colonization of the host organ. Exosomes
Available online xxx
influence every step of this cascade and can be targeted by oncological treatment. This review highlights
the role of exosomes in the various steps of the metastatic cascade and how exosome dependent pathways
Keywords:
can be targeted as therapeutic approach or used for liquid biopsies.
Tumor derived exosomes
© 2017 Elsevier Ltd. All rights reserved.
Premetastatic niche
Organotropic metastasis
EMT
Cancer-associated fibroblasts

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Biogenesis and morphology of exosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Morphology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Biogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3. Exosomal content . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.4. Biological functions and effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.5. Isolation methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Initiation of metastasis in the host organ: exosomes as mediators in EMT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1. EMT and migration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.2. EMT and invasion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.3. Exosomes and CAFs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Engraftment of metastatic cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.1. Exosomes and MErT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.2. Exosomes and neoangiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.3. Exosomes in organotropic metastatic growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.4. TDEs and organotropism in malignant melanoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.5. TDEs and organotropism in pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Immune-modulating effects of exosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1. Maintenance of metastatic cells in the blood circulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.2. Maintenance of metastatic cells in the host organ: TDEs and cellular immune response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.3. Maintenance of metastatic cells in the host organ:TDEs and humoral immune response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

∗ Corresponding author at: Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology,
Medical University of Innsbruck, Anichstr. 35, A-602, Innsbruck, Austria.
E-mail addresses: Ira.Skvortsova@i-med.ac.at, Ira.Skvortsova@Tirol-kliniken.at (I.-I. Skvortsova).

http://dx.doi.org/10.1016/j.semcancer.2017.02.006
1044-579X/© 2017 Elsevier Ltd. All rights reserved.

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
2 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

5.4. TDEs and cancer-promoting proinflammatory effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


6. Exosomes as a source for biomarkers predicting metastatic spread . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1. Exosomal protein markers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.2. Exosomal miRNA as biomarker . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Therapeutic approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.1. Exosomes as cancer vaccination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.2. M-Trap . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.3. Exosomes as chemotherapeutic agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction receptor-ligand interaction or by direct fusion and dependent on


microenvironmental pH [14,15] (Fig. 2).
Most studies of the pathogenesis of metastasis focus on genetic
or phenotypic changes of the cancer cell itself, however, there is
growing evidence that cancer cells communicate with each other
and the surrounding stroma leading to metastasis [1]. 2.3. Exosomal content
Metastasis is a multistep process including invasion of the tumor
cell through the basal membrane and into blood vessels, survival Exosomes contain cytosolic proteins or proteins of the
in the blood circulation, attachment to the blood vessels, extrava- plasma/endosome membrane while proteins of the nucleus, mito-
sation and at last colonization and growth in the host organ [2]. chondria, endoplasmatic reticulum or golgi-appartus are absent
Cells can communicate with various types of extracellular mem- [16]. The sorting of proteins into exosomes is an area of current
brane vesicles. These include exosomes, proteasomes, apoptotic research and is at least in part dependent on protein ubiquitylation
blebs and microvesicles [3]. Since exosomes can promote a vari- and the ESCRT (endosomal sorting complex required for transport
ety of intratumoral biological processes, it seems likely that they machinery) [3,17,18] (Fig. 1). The ESCRT form small necks that occur
can also influence metastatic properties of malignant tumors. The at the plasma membrane during exosome formation. They contain
following review focuses on the role of exosomes in this multistep filaments, flat spirals, tubes and conical funnels that are thought to
metastatic process. direct membrane remodeling. Their assembly and disassembly is
ATP-dependent [19].
Exosomes contain a high amount of transport proteins like
2. Biogenesis and morphology of exosomes
tubulin, actin and actin-binding molecules [16] as well as several
proteins associated with specific functions of secreting cells [8].
2.1. Morphology
Nearly all exosomes carry MHC class I- molecules [20] and heat
shock protein (HSP), especially HSP 70 and HSP 90 [8,16]. HSP
Trams and co-authors first described exosomes in 1981
70 and HSP 90 are involved in antigen presentation and can bind
as microvesicles containing 5‘-nucleotidase activity which are
antigenic peptides to MHC class I-molecules [8]. Exosomes carry
released by neoplastic cell lines [4]. A few years later their endo-
high concentrations of tetraspanins – including CD9, CD63, CD81
cytic origin was proven [5]. Exosomes can be identified by a few
and CD82–that are rarely present in other microvesicles and are
characteristics. They have a size between 40 and 100 nm and are
involved in antigen presentation and adhesion through interaction
secreted after fusion of multivesicular endosomes with the cell sur-
with other transmembrane proteins. CD9 and CD82 inhibit migra-
face or by direct budding from the plasma membrane [6]. They
tion and invasion of tumor cells in vitro and in vivo by interaction
are round or have a cup shaped morphology and have a density
with integrins [21]. To date more than 41 860 different proteins
between 1.13 and 1.19 g/ml [3]. Exosomes can be identified by their
have been reported as cargo of exosomes and are listed in online
unique protein and lipid composition and double lipid layer due to
databases like Exocarta (http://www.exocarta.org) and Visclepedia
their origin in the fusion of late endocytic compartments with the
(http://www.microvesicles.org).
plasma membrane [7].
In addition to proteins, exosomes contain lipids, particularly
raft-lipids like ceramides, sphingolipids, cholesterol and glyc-
2.2. Biogenesis erophospholipids as well as micro-RNA, mRNA and DNA enabling
cells to exchange genetic information [22].
Exosomes ori ginate in a reverse budding event, causing the Mi-RNAs are small non-coding RNAs that bind within the 3 -UTR
vesicles to carry cytosol and expose the extracellular domain of 
(3 -untranslated region) of mRNAs leading to destabilisation and
surface receptors [8]. They constitutively fuse with the plasma fragmentation of these RNAs and consequently to reduced expres-
membrane either after Ca++ -dependent activation or after acti- sion of the encoded protein [23,24]. That miRNA is the major RNA
vation of Rab-GTPases [9]. Rab25 regulates exosome binding to component of exosomes and was first discovered at the Univer-
and tethering with the plasma membrane and Rab27b exosome sity of Gotteburg by the scientific group around Prof. J. Lotvall in
release [10,11] (Fig. 1). The increased secretion of exosomes by can- 2007 [25,26]. This mode of transportation makes the miRNA resis-
cer cells is induced by overexpression of Rab3D (a small GTPase, tant of degradation through extracellular ribonculeases [27]. The
[12]. Release of exosomes can also be induced by diverse signalling miRNA transported in TDEs (tumor derived exosomes) reflects the
pathways including the activation of the Wnt pathway, which is dysregulated miRNA profile of the cancer cells and can be trans-
especially important in deregulated exosome secretion in cancer ported to different cell types or to the pre-metastatic niche enabling
cells [13]. The acidic microenvironment around tumors stimulates wide spread influence on the gene expression of target cells [28].
the release of exosomes and enhances their cell fusion capabili- Despite solely transporting miRNA, exosomes are able to process
ties [14]. The uptake of exosomes is accomplished via endocytosis, pre-miRNAs to mature-miRNAs, allowing direct interaction with

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx 3

Fig. 1. Exosomes originate in a reverse budding event, causing the vesicles to carry cytosol and expose the extracellular domain of surface receptors [8]. They form by invagi-
nation and budding from the plasma membrane, meaning that they are of endocytic origin. Intracellular exosomes are present in multivesicular bodies. The sorting of proteins
into exosomes is dependent on protein ubiquitylation and the ESCRT (endosomal sorting complex required for transport machinery) [3,17,18]. Exosomes constitutively fuse
with the plasma membrane either after Ca++ -dependent activation or after activation of Rab-GTPases [9]. Rab25 regulates exosome binding to and tethering with the plasma
membrane and Rab27b exosome release [10,11].

Fig. 2. Release of exosomes by donor cells can be induced by diverse signalling pathways and occurs in a reversed budding event as displayed in Fig. 1 [13]. The uptake of
exosomes is accomplished via endocytosis, receptor-ligand interaction or by direct fusion and dependent on microenvironmental pH [14,15].

the transcriptome of the recipient cell without the need of further ment and remodelling and cell-to-cell spread of infectious agents
processing these miRNAs [29]. like the HI-virus. The release of exosomes discards membrane pro-
The second major RNA content of exosomes are lnRNAs (long teins that have become useless as an alternative to the classical
non-coding RNAs, >200 bp) that interfere with gene expression in lysosomal degradation [31].
various ways such as direct annealing with genomic DNA or by In cancer development exosomes are further described as
modifying histone complexes [30] (Fig. 3) (Table 1). functional mediators of tumor-stroma interaction [32]. This inter-
cellular communication is known to be involved in various
pathophysiological processes including migration, treatment resis-
2.4. Biological functions and effects tance and metastasis [33].
Exosomes enable intercellular communication without direct
Exosomes promote intercellular communication and are cell-cell contact. They may confer epigenetic changes in the recip-
involved in various physiological as well as pathological processes, ient cells by transportation of microRNAs.
such as antigen presentation, immune regulation, tissue develop-

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
4 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

Fig. 3. Exosomal content: Exosomes contain cytosolic proteins or proteins of the plasma/endosome membrane while proteins of the nucleus, mitochondria, endoplasmatic
reticulum or golgi-appartus are absent [16]. Furthermore they contain RNA, especially mi-RNA, DNA and lipids.

Table 1 Before performing functional assays or downstream proteomic


Exosomal content.
or genomic approaches, purity of isolated exosomes should be
cytoskeletal proteins e.g. actin, tubulin assessed [35]. This can be done by using different biomarkers,
signal transduction molecules e.g. G-proteins which distinguish between exosomes and other extracellular vesi-
chaperones e.g. HSP70, HSP 90 cles [39]. Another criticism is that studies with purified exosome
membrane transport/fusion e.g. Rab 25, Rab 27b, annexins isolation may not be physiological relevant due to the higher dilu-
antigen presentation e.g. MHC-I, MHC-II tion of exosomes in body fluids. Most exosome studies are either
adhesion molecules e.g. tetraspanins (CD9, CD63,
in vitro analyses or mouse models, while clinical studies are rare
CD81, CD82), integrins
ESCRT components [37].
other cytosolic proteins e.g. histones, ribosomal
proteins, proteasomes
other transmembrane proteins e.g. LAMPs
lipids e.g. sphingomyeline, ceramid,
3. Initiation of metastasis in the host organ: exosomes as
glycerophospholipids,
cholesterol mediators in EMT
nucleic acids mi-RNA, lnRNA, DNA
The invasion metastasis cascade describes the functional pro-
cess of building macrometastases in distant organs. Tumor cells
2.5. Isolation methods loose their adhesion to the stroma and migrate into the blood
stream. After reaching their pre-metastatic niche they either enter a
Differential ultracentrifugation is the current reference method dormant state as DTCs (dormant tumor cells) or proliferate to form
for exosome isolation. It consists of multiple centrifugation steps multicellular micrometastases, from which finally macrometas-
with increasing centrifugal strength to sequentially pellet cells tases arise. Metastasis is an inefficient process; only 0.01% of the
(300g), microvesicles (10,000g) and exosomes (100,000g) [34,35]. tumor cells that reach the blood stream are able to proliferate in
It is fast and simple but the exosome pellet contains large amounts distant organs.
of contaminating soluble proteins and non-exosomal particles EMT (epithelial to mesenchymale transition) frequently initi-
[36,37]. ExoQuick precipitation is also based on centrifugation, ates the metastatic process. EMT means that epithelial tumor cells
but before ultracentrifugation exosomes are chemically precipi- acquire mesenchymal characteristics under the influence of CAFs
tated with ExoPrep Solution (Hansa Bio Med, Tallinn, Estonia). This (cancer-associated fibroblasts) in the tumor stroma. Under EMT,
results in similar advantages and disadvantages [34]. ExoQuick is tumor cells loose their polarity and cell-cell junctions and turn to a
a proprietary polymer that precipitates exosomes. Currently, the low proliferation state with increased migratory and invasion capa-
purest exosome preparation can be obtained by density-gradient bilities [40]. Molecular markers of EMT are a loss of E-cadherin as
isolation methods using sucrose or iodixanol (OptiPrepTM , Sigma- epithelial marker and an increased expression of vimentin as mes-
Aldrich). Iodixanol is capable of forming iso-osmotic solutions at enchymal marker as well as a spindle-like cell shape [1,41]. These
all densities, thus better preserving the size of the vesicles in the morphological changes are strongly associated with activation of
gradient. These procedures are more time consuming and costly the Snail (zinc finger proteins Snail and Slug), Zeb (zinc finger and
[35,38]. Another purification method is microfluidic immunoiso- homeodomain proteins Zeb 1 and 2) and Twist (basic helix-loop-
lation, where serum flows through microchambers, which contain helix proteins E12, E47, Twist1, Twist 2 and Id) pathways [40]. Once
antibodies against exosomal markers like EpCAM or CD63. A key the tumor cells have reached the distant pre-metastatic niche the
limitation is that this method focuses only on the exosomes that reversed process takes place. This so-called MErT (mesenchymal to
express the target surface marker and will therefore fail to identify epithelial reverting transition) returns tumor cells to a high prolif-
other subpopulations of exosomes [35,37]. erative state and enables formation of macrometastases (Fig. 4).

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx 5

Fig. 4. EMT and MErT.


Epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial reverting transition (MErT) are two reversible events, which merge into one another. EMT is
characterised by an increased expression of mesenchymale markers like vimentin and a decreased expression of epithelial markers like E-cadherin and ␤-catenin. MErT is
characterised by an increased expression of these epithelial markers and a constant expression of the mesenchymal marker vimentin.

3.1. EMT and migration miR-105 in the serum of patients with breast cancer is a prognostic
marker for the later development of metastasis and serum miR-105
Recent investigations suggest a major role of exosomes in EMT. level correlates with the miR-105 level in the primary tumor [28].
Exosomes from EBV infected nasopharyngeal carcinoma cells con- Blocking of astrocyte-exosomes inhibits brain metastasis in vivo.
tain high levels of LMP1 and HIF1␣ (hypoxia-inducible factor 1␣). This effect is dependent on exosomal miRNA, which blocks PTEN in
HIF1␣ interacts with the Snail pathway and increases Twist expres- brain-metastatic tumor cells [57].
sion inducing EMT in nasopharyngeal carcinoma cells [42]. This Exosomal miR-21 level in the serum from patients with
results in a more invasive phenotype in recipient cells. The increase esophageal cancer correlates with cancer recurrence and metas-
in HIF1␣ in nasopharyngeal carcinoma depends on the expression tasis [58] and was described as a marker of tumor recurrence in
of LMP1 (latent membrane protein1), an important oncogene of lung cancer and malignant melanoma [59,60]. MiR-21 promotes
EBV (Epstein-Barr-virus). LMP1 decreases the lysosomal degrada- migration and invasion of recipient cells in esophageal and oral
tion of HIF1␣ by stabilizing seven in absentia homolog and causing squamous cell carcinoma [58,61].
activation of EMT pathways [43,44].
Treatment of urothelial cells with exosomes isolated from 3.3. Exosomes and CAFs
muscle-invasive bladder cancer cells resulted in an EMT like
phenotype and increased migratory and invasive features. These TDEs are not only able to force other cancer cells to acquire a
exosomes induced the expression of vimentin, Snail and Twist in mesenchymal phenotype, but are also able to convert mesenchy-
urothelial cell lines while decreasing E-cadherin and ␤-catenin mal stem cells into cancer-associated fibroblasts (CAFs) with an
expression via the TGF- ␤1 pathway. In contrast, exosomes from increased expression of ␣-SMA (␣-smooth muscle actin) [62]. These
embryonic kidney cells failed to induce these EMT-like pheno- CAFs reciprocally cross talk with cancer cells and are characteris-
typic changes, suggesting that not exosomes in general but tumor tically surrounded by a layer of hyaluronic acid, a condition which
derived exosomes (TDEs) in particular are able to induce EMT normal fibroblasts lack. TDEs are able to induce such alterations
and enhance migratory activity [45,46]. This pro-migratory, EMT- in the pericellular microenvironment [62]. CAFs in return stimu-
inducing effect of TDEs was also observed in glioblastoma cell late EMT by secretion of TGF-␤1 (transforming growth factor ␤)
lines, lung carcinoma cells and an in vivo model of gastric cancer and consequently activate the TGF-␤1-SMAD (small mother against
[47–49]. Exosomal miR-23a supports the EMT-promoting effect of decapentaplegic protein) signalling pathway. Interestingly, soluble
TGF-␤1 by inhibiting E-cadherin synthesis in lung carcinoma and TGF-␤1 alone is not able to drive stroma differentiation to a cancer-
melanoma cells [50,51]. MiR-191 and let7a are two further miRNA associated phenotype. Exosome-depleted cancer cells fail to gain a
modulators of EMT found in exosomes of patients with malignant stroma-mediated growth advantage in vivo and the TGF-␤1 effect
melanoma, gastric cancer and colorectal cancer [52–54]. Further on stroma differentiation is abrogated by blocking of exosomes,
mi-RNA transported in exosomes with pro- and anti-oncogenic even if the TGF-␤1 levels in the tumor stroma remain constant [63].
effects is displayed in Table 1 [55]. These results suggest that the biophysical format of TGF-␤1 from
TDEs from tumor cells who have undergone EMT can in turn exosomes drives stroma-modifying effects [63].
stimulate neighbouring cells to acquire EMT like features, creating CAFs themselves produce exosomes, which stimulate the Wnt-
a synergistic effect [56]. pathway in breast cancer cells and enhance their migratory
capabilities [64].
3.2. EMT and invasion CAFs produce tracks in the extracellular matrix with the help
of enzymes like matrix-metalloproteinases – e.g. MMP 13 – and
Apart from the pro-migratory effects of exosomes through create tunnels through which the tumor cells can follow [65]. MMP-
EMT-related pathways, TDEs directly influence invasion into blood 13 expression is a prognostic marker for invasiveness and poor
vessels by the secretion of miR-105. This process destroys tight clinical outcome in various cancer types such as head and neck can-
junctions via interaction with ZO-1 in breast cancer models cer [66]. Exosomal MMPs stimulate tumor metastasis in sarcomas
suspending a natural barrier against metastasis [28]. Exosomal [67] and exosomal CD81 functions in the context of CAF induced

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
6 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

breast cancer cell mobility [68]. Exosomal CD151 supports matrix in vitro in malignant melanoma [85]. TDEs promote education and
degradation and mobility in an in vitro model of pancreatic adeno- mobilization of BMDCs that support tumor vasculogenesis, inva-
carcinoma [69]. It has been demonstrated that CAFs and tumor cells sion and metastasis. TDEs recruit BMDCs through upregulation of
jointly invade blood vessels and implant themselves in metastatic proinflammatory molecules and further enhance vascular leakiness
sites [70]. at the metastatic sites [85]. TDE miR-122 downregulates pyru-
vate kinase in non-malignant cells from the premetastatic niche by
4. Engraftment of metastatic cells impairing glucose utilization in non-malignant cells and increasing
the nutritional supply of the cancer cells [86,87] (Fig. 5).
4.1. Exosomes and MErT These findings raise the question of how exosomes are directed
to the specific metastatic organs enabling organotropic metastatic
After metastatic cells have colonized in a secondary organ growth. Melanoma exosomes are home to sentinel lymph nodes
they often display epithelial features, suggesting at least partial in vivo and recruit melanoma cells [88]. Exosomes from breast can-
mesenchymal-epithelial reverting transition (MErT). While mes- cer cells move to lung tissue in orthotopic nude mouse models [89].
enchymal markers like vimentin persist, they often express higher Hohsino et al. described in 2015 that TDEs are sufficient to redirect
amounts of E-cadherin than the primary tumor. MErT returns the metastatic tumor cells that normally lack the ability of organ spe-
cancer cells to a highly proliferative but less migratory phenotype, cific metastasis, meaning that the host organ is prepared prior to
enabling the development of macrometastasis [71]. This is a con- metastatic dissemination of the cancer cells [90]. Exosome biodis-
sequence of an increased membranous ␤-catenin expression in the tribution matched the organotropic metastatic spread in vitro in cell
metastatic cells, which results in stable adherent junctions. Inter- lines from various types of cancer such as breast and pancreatic can-
estingly, E-cadherin expression decreases with increasing size of cer. The biodistribution of exosomes is at least in part mediated by
the metastasis, suggesting that MErT itself is reversible and the cells integrins. ITG␤4 (integrin beta 4) promotes lung tropism and ITG␤5
can re-enter a mesenchymal phenotype after EMT [72]. In prostate (integrin beta 5) promotes exosome adhesion in the liver. Accord-
cancer patients these findings were confirmed in haematogenous ingly, increased ITG␤4 levels were expressed in exosomes from the
metastasis but not in metastatic cells in the surrounding lymph serum of patients with lung metastasis (regardless of the primary
nodes, where the tumor cells resemble the E-cadherin expression tumor site) and increased ITG␤5 levels were found in the serum
of the primaries [73] (Fig. 4). of patients with liver metastasis (with pancreatic carcinoma) [90]
(Fig. 5).
4.2. Exosomes and neoangiogenesis
4.4. TDEs and organotropism in malignant melanoma
After the metastasis reaches a critical size, neoangiogenesis
has to be stimulated to avoid cancer necrosis. Neoangiogenesis is Exosome protein concentrations in the plasma of patients with
important for the nutrition and oxygen supply of the tumor and stage IV Melanoma were significantly higher than in the plasma
acts as entry point for metastatic cells into systemic circulation of patients with lower stages or healthy individuals. Accordingly,
[74]. CAFs stimulate neoangiogenesis via secretion of exosomal individuals with stage IV disease with lower exosome concentra-
SDF-1 (stromal cell derived factor-1) [63,75]. The released SDF-1 tions in the plasma (<50 ␮g/ml) had a survival advantage over
is responsible for the recruitment of endothelial progenitor cells ones with higher concentrations. Exosomes isolated from highly
by chemotaxis [76,77]. Similar results where found after treatment metastatic melanoma cell lines achieve organ tropism of melanoma
of umbilical cord mesenchymal stem cells with gastric-cancer cells cells by harbouring in the interstitial space of the lungs, bone mar-
derived exosomes or with exosomes from hepatic cancer cells [78]. row, kidney, liver and spleen – the organotropic sites for malignant
TDEs stimulate endothelial progenitor cells to form tube-like struc- melanoma – but do not remain in the blood circulation. They fur-
tures [79]. Induction of neoangiogenesis is not only due to the ther enhance the permeability of the lung endothelium facilitating
exosome’s transport of paracrine signalling factors but also due tumor cell invasion. These exosomes enhance the expression of
to direct transport of relevant mRNA to the surrounding stroma, genes like HSPs (especially HSP 90 and 70), S100A8 and S100A9
influencing gene expression directly. In this process the transport which are involved in extracellular matrix remodelling and inflam-
of EGFR (epidermal growth factor receptor)-mRNA [79] and mRNA mation, annexin A6, CD44, especially CD44v6–a splice variant
related with cell cycle progression of endothelial cells [80] is impor- strongly associated with metastasis promotion [91–93] – and the
tant. Met oncoprotein. BMDCs expressed higher levels of Met, which
is a key player in migration, invasion, angiogenesis and bone
4.3. Exosomes in organotropic metastatic growth marrow cell mobilization via the mTOR (mammalian target of
rapamycin) and MAPK (mitogen activated protein kinase) path-
Organotropic metastasis means that the side of metastasis is way [85]. Exosome educated bone marrow in a murine xenograft
not chosen randomly but is a rather a consequence of intricate model increased metastatic spread three fold and the number of
tumor-stroma interaction in the host organ [81]. This hypothesis metastasis-associated BMDCs was higher in the typical metastatic
was first postulated in 1889 by Paget as “soil and seed hypothesis”, areas (Fig. 5).
which means that circulating tumor cells form metastasis only if
the seed (tumor cell) and host (organ) are compatible [82]. Fidler 4.5. TDEs and organotropism in pancreatic cancer
et al. supported this hypothesis in 1973 and showed that metastatic
cancer cells derived from a particular tumor site display enhanced Similar observations were made in a mouse model for pan-
metastatic ability to specific organs, independent of the anatomy creatic ductal adenocarcinoma, where TDEs support engraftment
of blood and lymphatic vessels that drain the primary tumor site of metastatic cells in the liver by interaction with Kupffer-cells
[83]. A pre-metastatic niche formation is required for tumor cells [94]. Uptake of exosomes by Kupffer-cells causes activation of pro-
to engraft a distant organ. This pre-metastatic niche is formed with inflammatory and fibrotic pathways leading to the formation of a
the help of bone marrow derived cells (BMDCs, e.g. haematopoi- pre-metastatic niche through MIF (migration inhibitory factor) sig-
etic cells), endothelial progenitor cells and mesenchymal cells [84]. nalling, which supports the production of TGF-␤1. TGF-␤1 in turn
Peinado et al. described that this pre-metastatic niche formation is supports the expression of fibronectin, which attracts BMDCs to the
in part signalled through TDEs, which influence BMDCs in vivo and liver stroma in mice. Significant higher levels of MIF where found

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx 7

Fig. 5. Organotropic metastatic growth and premetastatic niche formation.


Tumor derived exosomes (TDEs) are distributed to the specific metastatic sites via integrins. ITG␤4 promotes lung tropism and ITG␤5 promotes liver tropism. TDEs prepare
the metastatic organ to form a premetastatic niche, with the help of immigrating bone marrow derived progenitor cells (BMDCs). TDEs promote education and mobilization
of BMDCs that support tumor vasculogenesis, invasion and metastasis. TDEs recruit BMDCs through upregulation of proinflammatory molecules, like miR-122, S100 and
fibronectin, and enhance vascular leakiness at the metastatic sites [85]. After premetastatic niche formation cancer cells can invade the distant organ, begin colonialization
and form organspecific metastasis.

in the serum of patients with pancreatic ductal adenocarcinoma adhesion to these metastatic cells shield them from NK cell activ-
with liver metastasis and progressive disease than in patients with ity and enables survival in the blood stream [105]. One mechanism
full remission, suggesting MIF as prognostic marker in pancreatic to abrogate NK cell activity is by coating tumor cells with fibrin,
carcinoma [94]. High levels of TGF-␤1, a down stream mediator of reducing recognition by NK cells [101]. A second mechanism is that
MIF, are associated with a poor prognosis in pancreatic ductal ade- exosomes from hypoxic tumor cells can impair NK cell function by
nocarcinoma [95]. TGF-␤-inhibitors have been recently evaluated secretion of TGF-ß1, which leads to a decreased expression of NK
with promising results in phase-I and phase-II clinical trials [96]. cell activating receptor (NKG2D) [106,107]. This interaction leads
to activation of the EMT-associated TGF-␤1 pathway, which plays
5. Immune-modulating effects of exosomes an important role in metastasis as mentioned before [108].

5.1. Maintenance of metastatic cells in the blood circulation 5.2. Maintenance of metastatic cells in the host organ: TDEs and
cellular immune response
To survive in the blood circulation and to successfully prolif-
erate in the host organ, metastatic cells need to escape immune After arrival at the pre-metastatic niche, TDEs establish an
surveillance. Platelets are effector cells in the primary haemosta- immunosuppressive environment to maintain tumor cell prolif-
sis but they are also immune competent. The first link between eration. Both, the cellular immune response and the humoral
platelets and cancer was observed in 1865 by Trousseau, who immune response can be abjured by TDEs. TDEs reduce cellu-
diagnosed excessive blood clotting in cancer patients [97]. Platelet lar immune response by inhibiting effector cells and stimulation
derived exosomes contain P-selectin and GP IIb-IIIa (glycoprotein of regulatory T-cells. In an in vitro culture model of AML (acute
IIa-IIIB) and interact with endothelial cells, leukocytes and cancer myeloid leukaemia), TDEs decreased the count of CD8+-T-cells by
cells [98,99]. Platelets accompany cancer cells in the blood stream activation of Fas/FasL-mediated apoptosis [109,110]. They further
and enable their survival. They assist tumor cell adhesion at the promoted CD4+-T-cell proliferation and conversion into regulatory
vessel endothelium, especially via P-selectin, enabling extravasa- T-cells with increased expression of IL-10 (interleukin-10), TGF-␤1,
tion of tumor cells from the blood stream into the premetastatic CTLA-4 (cytotoxic lymphocyte antigen-4) and GrB (granzyme B).
niche [100–102]. Platelet depletion inhibits metastasis in mice, These mediators reduce cytotoxic activity of NK cells [111,112]. In
while reconstitution of these platelets restores metastatic activ- an in vitro model of nasopharyngeal cancer, immunosuppressive
ity [103]. Under normal circumstances, leukocytes and especially miRNAs (hsa-miR-24-3p, hsa-miR-891a, hsa-miR-106a-5p, hsa-
natural killer (NK) cells are found in close relation to metastatic miR-20a-5p, and hsa-miR-1908) were found in TDEs [113,114].
cells in the blood stream and eliminate these cells [104]. Platelet Myeloid-derived suppressor cells are immature myeloid cells with

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
8 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

the ability to suppress T-cell-activation [115]. TDEs activate human gates and other membranous particles is a prerequisite to identify
myeloid derived suppressor cells through HSP72/TLR-2 (toll-like exosome-specific biomarkers [35].
receptor 2) via the STAT3 pathway and IL-6 expression [116].
6.1. Exosomal protein markers
5.3. Maintenance of metastatic cells in the host organ:TDEs and
humoral immune response Exosomes from cancer cells have unique properties, suggesting
that they can be used as early biomarkers for cancer detec-
In addition to the effect of TDEs on cellular immune response, tion, relapse and metastatic spread. Glypican-1–carrying exosomes
TDEs also neutralize antitumor-antibodies. For the humoral were detected in the serum of 246 pancreatic cancer patients
immune response against cancer cells antibody-dependent cyto- with 100% specificity and 100% sensitivity compared to 20 healthy
toxicity (ADCC) is the major response mechanism and to a lesser donors and 37 patients with benign pancreatic lesion. This suggests
extent complement-dependent cytolysis. The activity of thera- it is a useful predictive biomarker to distinguish between benign
peutic antibodies is mainly dependent on ADCC [117]. In breast and malign pancreatic lesions. Further the Glypican-1–carrying
cancer cells it was demonstrated that TDEs express Her2 (human exosome concentration correlated with the tumor load allowing
epidermal growth factor receptor 2) and EpCAM (epithelial cell diagnosis of relapse after surgery or metastatic spread [124]. These
adhesion molecule) antigens, which bind and neutralize anti- Glypican-1-carrying exosomes can be detected in the serum of
bodies interfering with ADCC of tumor cells expressing these patients with gliomas and breast cancer, too [124]. Another exam-
antigens. As a consequence, these TDEs may reduce the therapeu- ple for exosomes as predictive biomarkers is exosomal TM256
®
tic effect of Trastuzumab (Herceptin , mammalian Her2-antibody), (transmembrane protein 256) in urine of patients with prostate
which is a major therapeutic drug in metastatic breast cancer cancer, which has a very high sensitivity (94%) and 100% specificity
[118]. Accordingly, B-cell lymphoma cells secrete exosomes, which [126].
carry CD20, neutralizing the therapeutic effect of Rituximab (anti-
CD20-antibody) in vivo. Rituximab induces the formation of the 6.2. Exosomal miRNA as biomarker
membrane attack complex at the surface of TDEs. This reduces the
binding of Rituximab at the lymphoma cell surface, which results in Besides those proteins, exosomal miRNA is a useful source
cell lysis and death by disruption of the cell membrane integrity via for predictive biomarkers. Tumors display unique sets of miRNA
the membrane attack complex by forming transmembrane chan- expression profiles, suggesting that these profiles can be used for
nels. Interestingly, the treatment of lymphoma cells with Rituximab early diagnosis and diagnosis of relapse [127]. In prostatic car-
induced the secretion of exosomes carrying CD20. Depletion of the cinoma patients exosomal miR-141 and miR-375 were observed
patient plasma from exosomes increased the therapeutic effect of in higher concentrations in the plasma of patients with recurrent
Rituximab [119]. disease after radical prostatectomy compared to patients with non-
recurrent disease [128–130]. MiR-141 was described as biomarker
for the presence of metastasis [131].
5.4. TDEs and cancer-promoting proinflammatory effects
7. Therapeutic approaches
Inflammation stimulates tumor growth and prepares distant
sites of the body for hosting metastatic cells [84]. Besides suppres- Exosomes stimulate tumor growth, invasiveness and metas-
sion of antitumor immune response, TDEs create a protumorigenic tasis. Consequently, inhibiting TDEs as therapeutic approach
inflammatory response and support the establishment and main- in cancer treatment seems reasonable. The chemical inhibitor
tenance of a pre-metastatic niche. Tumor-associated macrophages GW4869 blocks neutral sphingomyelinase 2, which is the key reg-
support neoangiogenesis, invasion and matrix remodelling [120]. ulator of ceramide biosynthesis. Exosome release can be triggered
Increased density of macrophages in breast cancer histological by a ceramide dependent pathway, which means that GW4869 is
sections correlate with a poor prognosis [121]. TDEs stimulate able to inhibit TDE release in vitro [132].
macrophages by activating NF-␬B (nuclear factor-␬B). These effects
are mediated through toll-like receptors (TLRs) on the surface of 7.1. Exosomes as cancer vaccination
macrophages, which interact with TDEs, especially TLR-2 [122].
Activation of macrophages by TLRs (TLR-7 and −8) could be Besides this approach of using TDEs as target for therapy, puri-
achieved by binding of exosomal miRNAs (mi-R21, mi-R29a) [123]. fied exosomes have been suggested as immunotherapy. Exosomes
TDEs of pancreatic ductal adenocarcinoma cells interact with carry antigen loaded MHC class I molecules. Antigen-presenting
Kupffer-cells in the liver to promote pre-metastatic niche forma- cells (APC)-derived exosomes carry these MHC/tumor antigens
tion. MIF (macrophage migratory inhibition factor) promotes the complexes and present them to dendritic cells, which then mediate
release of TGF-␤1 by Kupffer-cells leading to increased fibronectin T-cell activation and T-cell dependent immune responses against
production by hepatic stellate cells. Fibronectin deposits in the liver tumor cells in vivo. Consequently, these exosomes can be used as
promote the arrest of bone marrow derived cells in the liver creat- cell-free anti-tumor vaccination [133]. APC-exosomes have some
ing a pre-metastatic niche [94]. limitations, as APCs are difficult to culture and specific tumor anti-
gens need to be transferred to APCs. They are also MHC-I dependent
6. Exosomes as a source for biomarkers predicting and consequently require MHC-I haplotype matching, which means
metastatic spread they have to be prepared for each patient individually [134]. In
contrast to APC-exosomes, TDEs carry shared tumor antigens and
Normal human blood contains about 1.5 billion exosomes per not only antigens specific to one tumor, so these shared tumor
ml and the blood of cancer patients contains more than twice antigens could probably provide cross-protection against various
this concentration [124]. Diseased organs and cancer cells generate cancer types. Furthermore, they have antitumor immunogenity
more exosomes than healthy tissue [125]. One limitation why these even without expressing MHC-I molecules, allowing creation of a
biomarkers have not found their way in the clinical routine right cell-free vaccine that does not need MHC-I matching resulting in
now is that the isolation is expensive and time consuming. A robust effective cancer immunization across tumor types and MHC haplo-
method with the ability to minimize co-isolating protein aggre- types [135]. One shared tumor antigen is hMUCI (human Mucin I),

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx 9

a large glycoprotein, which is associated with rapid tumor progres- References


sion and poor outcome. Exosomes can be designed to carry MUCI
and act as anti-cancer vaccination in vitro in an MHC-I independent [1] J. Dudas, Supportive and rejective functions of tumor stroma on tumor cell
growth, survival, and invasivity: the cancer evolution, Front. Oncol. 5 (2015)
manner against MUCI- expressing tumors [136]. 44.
Hsp70 can be induced in tumor cells and in their exosomes by [2] A.F. Chambers, A.C. Groom, I.C. MacDonald, Dissemination and growth of
heat treatment of the cells. Hsp70 stimulates monocytes and den- cancer cells in metastatic sites, Nat. Rev. Cancer 2 (8) (2002) 563–572.
[3] M. Simons, G. Raposo, Exosomes-vesicular carriers for intercellular
dritic cells and can amplify the immune response triggered by TDEs communication, Curr. Opin. Cell Biol. 21 (4) (2009) 575–581.
[134]. Hsp70 is able to directly activate NK (natural killer) cells [4] E.G. Trams, C.J. Lauter, N. Salem Jr., U. Heine, Exfoliation of membrane
and initiate NK-mediated apoptosis of tumor cells via secretion of ecto-enzymes in the form of micro-vesicles, Biochim. Biophys. Acta 645 (1)
(1981) 63–70.
granzyme B [137]. Elsner et al. reported that activation of NK cells [5] B.T. Pan, K. Teng, C. Wu, M. Adam, R.M. Johnstone, Electron microscopic
results in a decreased size of the primary tumor and metastasis in a evidence for externalization of the transferrin receptor in vesicular form in
murine melanoma model [138]. Andre et al. reported in 2002 that sheep reticulocytes, J. Cell Biol. 101 (3) (1985) 942–948.
[6] M. Santoni, F. Piva, M. Scarpelli, L. Cheng, A. Lopez-Beltran, F. Massari, et al.,
exosomes isolated from ascites from cancer patients activate den-
The origin of prostate metastases: emerging insights, Cancer Metastasis Rev.
dritic cells and in turn produce an antitumor T-cell-response [139]. 34 (4) (2015) 765–773.
Two phase-I-clinical trials following this approach showed promis- [7] S. Keller, M.P. Sanderson, A. Stoeck, P. Altevogt, Exosomes: from biogenesis
ing results in patients with stage III/IV malignant melanoma [140] and secretion to biological function, Immunol. Lett. 107 (2) (2006) 102–108.
[8] C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and
and in patients with non-small lung carcinoma [141]. function, Nat. Rev. Immunol. 2 (8) (2002) 569–579.
[9] E.J. Blott, G.M. Griffiths, Secretory lysosomes, Nat. Rev. Mol. Cell Biol. 3 (2)
(2002) 122–131.
7.2. M-Trap [10] C. Hsu, Y. Morohashi, S. Yoshimura, N. Manrique-Hoyos, S. Jung, M.A.
Lauterbach, et al., Regulation of exosome secretion by Rab35 and its
De la Fuente A. et al. proposed a very innovative approach of GTPase-activating proteins TBC1D10A-C, J. Cell Biol. 189 (2) (2010) 223–232.
[11] S.R. Pfeffer, Two Rabs for exosome release, Nat. Cell Biol. 12 (1) (2010) 3–4.
using exosomes for therapy [142]. They designed a so called M-Trap
[12] J. Yang, W. Liu, X. Lu, Y. Fu, L. Li, Y. Luo, High expression of small GTPase
for metastatic cells, which is composed of exosomes embedded in Rab3D promotes cancer progression and metastasis, Oncotarget 6 (13)
a 3-D scaffold, and implanted these M-traps into the peritoneum (2015) 11125–11138.
of mice with ovarian cancer, establishing an artificial premetastatic [13] E.J. Ekstrom, C. Bergenfelz, V. von Bulow, F. Serifler, E. Carlemalm, G.
Jonsson, et al., WNT5A induces release of exosomes containing
niche in these mice which competes with the natural peritoneum pro-angiogenic and immunosuppressive factors from malignant melanoma
for metastatic cells. These M-traps cause a significant survival cells, Mol. Cancer 13 (2014) 88.
advantage in a murine model [142]. [14] I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi, A. De Milito, et al.,
Microenvironmental pH is a key factor for exosome traffic in tumor cells, J.
Biol. Chem. 284 (49) (2009) 34211–34222.
7.3. Exosomes as chemotherapeutic agents [15] X. Zhang, X. Yuan, H. Shi, L. Wu, H. Qian, W. Xu, Exosomes in cancer: small
particle, big player, J. Hematol. Oncol. 8 (2015) 83.
[16] C. Thery, M. Boussac, P. Veron, P. Ricciardi-Castagnoli, G. Raposo, J. Garin,
In addition to the properties described above, exosomes or et al., Proteomic analysis of dendritic cell-derived exosomes: a secreted
synthetic exosomes like liposome preparations can also trans- subcellular compartment distinct from apoptotic vesicles, J. Immunol. 166
port classical chemotherapeutics − like Cisplatin (then called (12) (2001) 7309–7318.
[17] D.J. Katzmann, M. Babst, S.D. Emr, Ubiquitin-dependent sorting into the
Lipoplatin) and Doxorubicin − to the tumor cells. This technique multivesicular body pathway requires the function of a conserved
reduces the toxicity of the chemotherapeutic agents and should endosomal protein sorting complex, ESCRT-I, Cell 106 (2) (2001) 145–155.
be preferred against conventional chemotherapy [143,144]. Pacli- [18] F. Reggiori, H.R. Pelham, Sorting of proteins into multivesicular bodies:
ubiquitin-dependent and – independent targeting, EMBO J. 20 (18) (2001)
taxel packed into exosomes is 50 times more cytotoxic against 5176–5186.
drug-resistant lung cancer cells than conventional paclitaxel in vitro [19] J. Schoneberg, I.H. Lee, J.H. Iwasa, J.H. Hurley, Reverse-topology membrane
[145]. In addition to classic chemotherapeutics, exosomes can also scission by the ESCRT proteins, Nat. Rev. Mol. Cell Biol. 8 (2017) 5–17.
[20] N. Blanchard, D. Lankar, F. Faure, A. Regnault, C. Dumont, G. Raposo, et al.,
transport so-called biologicals or tumorsuppressive mi-RNA to the TCR activation of human T cells induces the production of exosomes bearing
tumor cells [146,147]. Finally, they can be used for gene therapy the TCR/CD3/zeta complex, J. Immunol. 168 (7) (2002) 3235–3241.
carrying wild-type p53 DNA to cancer cells [148]. [21] M. Zoller, Tetraspanins: push and pull in suppressing and promoting
metastasis, Nat. Rev. Cancer 9 (1) (2009) 40–55.
[22] R. Wubbolts, R.S. Leckie, P.T. Veenhuizen, G. Schwarzmann, W. Mobius, J.
8. Conclusion Hoernschemeyer, et al., Proteomic and biochemical analyses of human B
cell-derived exosomes: potential implications for their function and
multivesicular body formation, J. Biol. Chem. 278 (13) (2003) 10963–10972.
Exosomes are involved in every step of the metastatic process [23] G.A. Calin, C.M. Croce, MicroRNA signatures in human cancers, Nat. Rev.
from invasion of the tumor cells into the surrounding tissues of Cancer 6 (11) (2006) 857–866.
the primary organ to the engraftment in the metastatic organ. [24] M.V. Iorio, M. Ferracin, C.G. Liu, A. Veronese, R. Spizzo, S. Sabbioni, et al.,
MicroRNA gene expression deregulation in human breast cancer, Cancer
The increasing knowledge about the biological function of these Res. 65 (16) (2005) 7065–7070.
microvesicles provides new insight into cancer biology and deliv- [25] R.S. Redis, S. Calin, Y. Yang, M.J. You, G.A. Calin, Cell-to-cell miRNA transfer:
ers biomarkers for cancer progression and new treatment options. from body homeostasis to therapy, Pharmacol. Ther. 136 (2) (2012)
169–174.
Therefore, identifying new therapeutic strategies and acquiring a [26] H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall,
better understanding of the biological functions of exosomes in Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism
cancer will be of uttermost relevance in modern molecular oncol- of genetic exchange between cells, Nat. Cell Biol. 9 (6) (2007) 654–659.
[27] M.J. Shurtleff, M.M. Temoche-Diaz, K.V. Karfilis, S. Ri, R. Schekman, Y-box
ogy.
protein 1 is required to sort microRNAs into exosomes in cells and in a
cell-free reaction, eLife (2016) 5.
Funding [28] W. Zhou, M.Y. Fong, Y. Min, G. Somlo, L. Liu, M.R. Palomares, et al.,
Cancer-secreted miR-105 destroys vascular endothelial barriers to promote
metastasis, Cancer Cell 25 (4) (2014) 501–515.
This study was supported by Austrian Science Fund (FWF, [29] S.A. Melo, H. Sugimoto, J.T. O’Connell, N. Kato, A. Villanueva, A. Vidal, et al.,
P29457). Cancer exosomes perform cell-independent microRNA biogenesis and
promote tumorigenesis, Cancer Cell 26 (5) (2014) 707–721.
[30] J.E. Wilusz, H. Sunwoo, D.L. Spector, Long noncoding RNAs: functional
Conflict of interest statement surprises from the RNA world, Genes. Dev. 23 (13) (2009) 1494–1504.
[31] J.S. Schorey, S. Bhatnagar, Exosome function: from tumor immunology to
pathogen biology, Traffic 9 (6) (2008) 871–881.
The authors indicate no potential conflicts of interest.

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
10 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

[32] Y. Kang, Dissecting tumor-stromal interactions in breast cancer bone [58] J. Liao, R. Liu, Y.J. Shi, L.H. Yin, Y.P. Pu, Exosome-shuttling microRNA-21
metastasis, Endocrinol. Metab. 31 (2) (2016) 206–212. promotes cell migration and invasion-targeting PDCD4 in esophageal
[33] A. Subramanian, V. Gupta, S. Sarkar, G. Maity, S. Banerjee, A. Ghosh, et al., cancer, Int. J. Oncol. 48 (6) (2016) 2567–2579.
Exosomes in carcinogenesis: molecular palkis carry signals for the [59] R. Munagala, F. Aqil, R.C. Gupta, Exosomal miRNAs as biomarkers of
regulation of cancer progression and metastasis, J. Cell Commun. Signal. recurrent lung cancer, Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 37 (8)
(2016). (2016) 10703–10714.
[34] C. Thery, S. Amigorena, G. Raposo, A. Clayton, Isolation and Characterization [60] S.R. Pfeffer, K.F. Grossmann, P.B. Cassidy, C.H. Yang, M. Fan, L. Kopelovich,
of Exosomes from Cell Culture Supernatants and Biological Fluids. Current et al., Detection of exosomal miRNAs in the plasma of melanoma patients, J.
Protocols in Cell Biology. Chapter 3:Unit 3, 2006, pp. 22. Clin. Med. 4 (12) (2015) 2012–2027.
[35] J. Van Deun, P. Mestdagh, R. Sormunen, V. Cocquyt, K. Vermaelen, J. [61] L. Li, C. Li, S. Wang, Z. Wang, J. Jiang, W. Wang, et al., Exosomes derived from
Vandesompele, et al., The impact of disparate isolation methods for hypoxic oral squamous cell carcinoma cells deliver miR-21 to normoxic cells
extracellular vesicles on downstream RNA profiling, J. Extracell. Vesicles to elicit a prometastatic phenotype, Cancer Res. 76 (7) (2016) 1770–1780.
(2014) 3. [62] J. Webber, R. Steadman, M.D. Mason, Z. Tabi, A. Clayton, Cancer exosomes
[36] H. Im, H. Shao, Y.I. Park, V.M. Peterson, C.M. Castro, R. Weissleder, et al., trigger fibroblast to myofibroblast differentiation, Cancer Res. 70 (23) (2010)
Label-free detection and molecular profiling of exosomes with a 9621–9630.
nano-plasmonic sensor, Nat. Biotechnol. 32 (5) (2014) 490–495. [63] J.P. Webber, L.K. Spary, A.J. Sanders, R. Chowdhury, W.G. Jiang, R. Steadman,
[37] A. Thind, C. Wilson, Exosomal miRNAs as cancer biomarkers and therapeutic et al., Differentiation of tumour-promoting stromal myofibroblasts by
targets, J. Extracell. Vesicles 5 (2016) 31292. cancer exosomes, Oncogene 34 (3) (2015) 290–302.
[38] M. Dettenhofer, X.F. Yu, Highly purified human immunodeficiency virus type [64] V. Luga, J.L. Wrana, Tumor-stroma interaction: revealing fibroblast-secreted
1 reveals a virtual absence of Vif in virions, J. Virol. 73 (2) (1999) 1460–1467. exosomes as potent regulators of Wnt-planar cell polarity signaling in
[39] J. Kowal, G. Arras, M. Colombo, M. Jouve, J.P. Morath, B. Primdal-Bengtson, cancer metastasis, Cancer Res. 73 (23) (2013) 6843–6847.
et al., Proteomic comparison defines novel markers to characterize [65] J.E. Park, H.S. Tan, A. Datta, R.C. Lai, H. Zhang, W. Meng, et al., Hypoxic tumor
heterogeneous populations of extracellular vesicle subtypes, Proc. Natl. cell modulates its microenvironment to enhance angiogenic and metastatic
Acad. Sci. U. S. A. 113 (8) (2016) E968–77. potential by secretion of proteins and exosomes, Mole. Cell. Proteom.: MCP
[40] M. Diepenbruck, G. Christofori, Epithelial-mesenchymal transition (EMT) 9 (6) (2010) 1085–1099.
and metastasis: yes, no, maybe, Curr. Opin. Cell Biol. 43 (2016) 7–13. [66] Y. You, Y. Shan, J. Chen, H. Yue, B. You, S. Shi, et al., Matrix metalloproteinase
[41] T.B. Steinbichler, V. Metzler, C. Pritz, H. Riechelmann, J. Dudas, 13-containing exosomes promote nasopharyngeal carcinoma metastasis,
Tumor-associated fibroblast-conditioned medium induces CDDP resistance Cancer Sci. 106 (12) (2015) 1669–1677.
in HNSCC cells, Oncotarget 7 (3) (2016) 2508–2518. [67] L. Min, J. Shen, C. Tu, F. Hornicek, Z. Duan, The roles and implications of
[42] J.L. Hood, Melanoma exosome induction of endothelial cell GM-CSF in exosomes in sarcoma, Cancer Metastasis Rev. (2016).
pre-metastatic lymph nodes may result in different M1 and M2 macrophage [68] V. Luga, L. Zhang, A.M. Viloria-Petit, A.A. Ogunjimi, M.R. Inanlou, E. Chiu,
mediated angiogenic processes, Med. Hypotheses 94 (2016) 118–122. et al., Exosomes mediate stromal mobilization of autocrine Wnt-PCP
[43] M. Aga, G.L. Bentz, S. Raffa, M.R. Torrisi, S. Kondo, N. Wakisaka, et al., signaling in breast cancer cell migration, Cell 151 (7) (2012) 1542–1556.
Exosomal HIF1alpha supports invasive potential of nasopharyngeal [69] S. Yue, W. Mu, U. Erb, M. Zoller, The tetraspanins CD151 and Tspan8 are
carcinoma-associated LMP1-positive exosomes, Oncogene 33 (37) (2014) essential exosome components for the crosstalk between cancer initiating
4613–4622. cells and their surrounding, Oncotarget 6 (4) (2015) 2366–2384.
[44] S. Shi, Q. Zhang, Y. Xia, B. You, Y. Shan, L. Bao, et al., Mesenchymal stem [70] T.A. Brentnall, L.A. Lai, J. Coleman, M.P. Bronner, S. Pan, R. Chen, Arousal of
cell-derived exosomes facilitate nasopharyngeal carcinoma progression, cancer-associated stroma: overexpression of palladin activates fibroblasts to
Am. J. Cancer Res. 6 (2) (2016) 459–472. promote tumor invasion, PLoS One 7 (1) (2012) e30219.
[45] C.A. Franzen, R.H. Blackwell, V. Todorovic, K.A. Greco, K.E. Foreman, R.C. [71] A. Wells, C. Yates, Shepard CR: E-cadherin as an indicator of mesenchymal to
Flanigan, et al., urothelial cells undergo epithelial-to-mesenchymal epithelial reverting transitions during the metastatic seeding of
transition after exposure to muscle invasive bladder cancer exosomes, disseminated carcinomas, Clin. Exp. Metastasis 25 (6) (2008) 621–628.
Oncogenesis 4 (2015) e163. [72] Y. Chao, Q. Wu, M. Acquafondata, R. Dhir, A. Wells, Partial mesenchymal to
[46] L.J. Vella, The emerging role of exosomes in epithelial reverting transition in breast and prostate cancer metastases,
epithelial-mesenchymal-transition in cancer, Front. Oncol. 4 (2014) 361. Cancer Microenviron.: Off. J. Int. Cancer Microenviron. Soc. 5 (1) (2012)
[47] H.C. Christianson, K.J. Svensson, T.H. van Kuppevelt, J.P. Li, M. Belting, Cancer 19–28.
cell exosomes depend on cell-surface heparan sulfate proteoglycans for [73] J. Pontes-Junior, S.T. Reis, M. Dall’Oglio, L.C. Neves de Oliveira, J. Cury, P.A.
their internalization and functional activity, Proc. Natl. Acad. Sci. U. S. A. 110 Carvalho, et al., Evaluation of the expression of integrins and cell adhesion
(43) (2013) 17380–17385. molecules through tissue microarray in lymph node metastases of prostate
[48] D. Liu, C. Li, B. Trojanowicz, X. Li, D. Shi, C. Zhan, et al., CD97 promotion of cancer, J. Carcinog. 8 (2009) 3.
gastric carcinoma lymphatic metastasis is exosome dependent, Gastric [74] S. Vong, R. Kalluri, The role of stromal myofibroblast and extracellular
Cancer: Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 19 (3) matrix in tumor angiogenesis, Genes Cancer 2 (12) (2011) 1139–1145.
(2016) 754–766. [75] J.A. Cho, H. Park, E.H. Lim, K.W. Lee, Exosomes from breast cancer cells can
[49] M.A. Rahman, J.F. Barger, F. Lovat, M. Gao, G.A. Otterson, P. Nana-Sinkam, convert adipose tissue-derived mesenchymal stem cells into
Lung cancer exosomes as drivers of epithelial mesenchymal transition, myofibroblast-like cells, Int. J. Oncol. 40 (1) (2012) 130–138.
Oncotarget (2016). [76] A. Huang, J. Dong, S. Li, C. Wang, H. Ding, H. Li, et al., Exosomal transfer of
[50] M. Cao, M. Seike, C. Soeno, H. Mizutani, K. Kitamura, Y. Minegishi, et al., vasorin expressed in hepatocellular carcinoma cells promotes migration of
MiR-23a regulates TGF-beta-induced epithelial-mesenchymal transition by human umbilical vein endothelial cells, Int. J. Biol. Sci. 11 (8) (2015)
targeting E-cadherin in lung cancer cells, Int. J. Oncol. 41 (3) (2012) 869–875. 961–969.
[51] J. Kim, T.Y. Kim, M.S. Lee, J.Y. Mun, C. Ihm, S.A. Kim, Exosome cargo reflects [77] A. Orimo, P.B. Gupta, D.C. Sgroi, F. Arenzana-Seisdedos, T. Delaunay, R.
TGF-beta1-mediated epithelial-to-mesenchymal transition (EMT) status in Naeem, et al., Stromal fibroblasts present in invasive human breast
A549 human lung adenocarcinoma cells, Biochem. Biophys. Res. Commun. carcinomas promote tumor growth and angiogenesis through elevated
(2016). SDF-1/CXCL12 secretion, Cell 121 (3) (2005) 335–348.
[52] K. Ohshima, K. Inoue, A. Fujiwara, K. Hatakeyama, K. Kanto, Y. Watanabe, [78] J. Gu, H. Qian, L. Shen, X. Zhang, W. Zhu, L. Huang, et al., Gastric cancer
et al., Let-7 microRNA family is selectively secreted into the extracellular exosomes trigger differentiation of umbilical cord derived mesenchymal
environment via exosomes in a metastatic gastric cancer cell line, PLoS One stem cells to carcinoma-associated fibroblasts through TGF-beta/Smad
5 (10) (2010) e13247. pathway, PLoS One 7 (12) (2012) e52465.
[53] S. Tanaka, M. Hosokawa, K. Ueda, S. Iwakawa, Effects of decitabine on [79] J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves,
invasion and exosomal expression of miR-200c and miR-141 in et al., Glioblastoma microvesicles transport RNA and proteins that promote
oxaliplatin-resistant colorectal cancer cells, Biol. Pharm. Bull. 38 (9) (2015) tumour growth and provide diagnostic biomarkers, Nat. Cell Biol. 10 (12)
1272–1279. (2008) 1470–1476.
[54] D. Xiao, S. Barry, D. Kmetz, M. Egger, J. Pan, S.N. Rai, et al., Melanoma [80] B.S. Hong, J.H. Cho, H. Kim, E.J. Choi, S. Rho, J. Kim, et al., Colorectal cancer
cell-derived exosomes promote epithelial-mesenchymal transition in cell-derived microvesicles are enriched in cell cycle-related mRNAs that
primary melanocytes through paracrine/autocrine signaling in the tumor promote proliferation of endothelial cells, BMC Genom. 10 (2009) 556.
microenvironment, Cancer Lett. 376 (2) (2016) 318–327. [81] X. Lu, Y. Kang, Organotropism of breast cancer metastasis, J. Mammary
[55] T. Lopatina, C. Gai, M.C. Deregibus, S. Kholia, G. Camussi, Cross talk between Gland Biol. Neoplasia 12 (2–3) (2007) 153–162.
cancer and mesenchymal stem cells through extracellular vesicles carrying [82] S. Paget, The distribution of secondary growths in cancer of the breast. 1889,
nucleic acids, Front. Oncol. 6 (2016) 125. Cancer Metastasis Rev. 8 (2) (1989) 98–101.
[56] M. Rodriguez, J. Silva, A. Herrera, M. Herrera, C. Pena, P. Martin, et al., [83] I.J. Fidler, Selection of successive tumour lines for metastasis, Nature: New
Exosomes enriched in stemness/metastatic-related mRNAS promote Biol. 242 (118) (1973) 148–149.
oncogenic potential in breast cancer, Oncotarget 6 (38) (2015) [84] B. Psaila, D. Lyden, The metastatic niche: adapting the foreign soil, Nat. Rev.
40575–40587. Cancer 9 (4) (2009) 285–293.
[57] L. Zhang, S. Zhang, J. Yao, F.J. Lowery, Q. Zhang, W.C. Huang, et al., [85] H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, G.
Microenvironment-induced PTEN loss by exosomal microRNA primes brain Moreno-Bueno, et al., Melanoma exosomes educate bone marrow
metastasis outgrowth, Nature 527 (7576) (2015) 100–104. progenitor cells toward a pro-metastatic phenotype through MET, Nat. Med.
18 (6) (2012) 883–891.

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx 11

[86] M.Y. Fong, W. Zhou, L. Liu, A.Y. Alontaga, M. Chandra, J. Ashby, et al., [113] D. Mrizak, N. Martin, C. Barjon, A.S. Jimenez-Pailhes, R. Mustapha, T. Niki,
Breast-cancer-secreted miR-122 reprograms glucose metabolism in et al., Effect of nasopharyngeal carcinoma-derived exosomes on human
premetastatic niche to promote metastasis, Nat. Cell Biol. 17 (2) (2015) regulatory T cells, J. Natl. Cancer Inst. 107 (1) (2015) 363.
183–194. [114] S.B. Ye, Z.L. Li, D.H. Luo, B.J. Huang, Y.S. Chen, X.S. Zhang, et al.,
[87] N. Kosaka, Decoding the secret of cancer by means of extracellular vesicles, Tumor-derived exosomes promote tumor progression and T-cell
J. Clini. Med. 5 (2) (2016). dysfunction through the regulation of enriched exosomal microRNAs in
[88] J.L. Hood, R.S. San, S.A. Wickline, Exosomes released by melanoma cells human nasopharyngeal carcinoma, Oncotarget 5 (14) (2014) 5439–5452.
prepare sentinel lymph nodes for tumor metastasis, Cancer Res. 71 (11) [115] S. Nagaraj, D.I. Gabrilovich, Regulation of suppressive function of
(2011) 3792–3801. myeloid-derived suppressor cells by CD4+ T cells, Semin. Cancer Biol. 22 (4)
[89] A. Suetsugu, K. Honma, S. Saji, H. Moriwaki, T. Ochiya, R.M. Hoffman, (2012) 282–288.
Imaging exosome transfer from breast cancer cells to stroma at metastatic [116] F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard, J.P. Remy-Martin,
sites in orthotopic nude-mouse models, Adv. Drug Deliv. Rev. 65 (3) (2013) et al., Membrane-associated Hsp72 from tumor-derived exosomes mediates
383–390. STAT3-dependent immunosuppressive function of mouse and human
[90] A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic myeloid-derived suppressor cells, J. Clin. Invest. 120 (2) (2010) 457–471.
Mark, et al., Tumour exosome integrins determine organotropic metastasis, [117] R.A. Clynes, T.L. Towers, L.G. Presta, J.V. Ravetch, Inhibitory Fc receptors
Nature 527 (7578) (2015) 329–335. modulate in vivo cytotoxicity against tumor targets, Nat. Med. 6 (4) (2000)
[91] Y.K. Chan, H. Zhang, P. Liu, S.W. Tsao, M.L. Lung, N.K. Mak, et al., Proteomic 443–446.
analysis of exosomes from nasopharyngeal carcinoma cell identifies [118] C. Battke, R. Ruiss, U. Welsch, P. Wimberger, S. Lang, S. Jochum, et al., Tumour
intercellular transfer of angiogenic proteins, Int. J. Cancer 137 (8) (2015) exosomes inhibit binding of tumour-reactive antibodies to tumour cells and
1830–1841. reduce ADCC, Cancer Immunol. Immunother.: CII 60 (5) (2011) 639–648.
[92] T. Jung, D. Castellana, P. Klingbeil, I. Cuesta Hernandez, M. Vitacolonna, D.J. [119] T. Aung, B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann, M. Lahmann, et al.,
Orlicky, et al., CD44v6 dependence of premetastatic niche preparation by Exosomal evasion of humoral immunotherapy in aggressive B-cell
exosomes, Neoplasia 11 (10) (2009) 1093–1105. lymphoma modulated by ATP-binding cassette transporter A3, Proc. Natl.
[93] Z. Wang, A. von Au, M. Schnolzer, T. Hackert, M. Zoller, CD44v6-competent Acad. Sci. U. S. A. 108 (37) (2011) 15336–15341.
tumor exosomes promote motility, invasion and cancer-initiating cell [120] B.Z. Qian, J.W. Pollard, Macrophage diversity enhances tumor progression
marker expression, Oncotarget (2016). and metastasis, Cell 141 (1) (2010) 39–51.
[94] B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, et al., [121] L.S. Ojalvo, C.A. Whittaker, J.S. Condeelis, J.W. Pollard, Gene expression
Pancreatic cancer exosomes initiate pre-metastatic niche formation in the analysis of macrophages that facilitate tumor invasion supports a role for
liver, Nat. Cell Biol. 17 (6) (2015) 816–826. Wnt-signaling in mediating their activity in primary mammary tumors, J.
[95] M. Javle, Y. Li, D. Tan, X. Dong, P. Chang, S. Kar, et al., Biomarkers of TGF-beta Immunol. 184 (2) (2010) 702–712.
signaling pathway and prognosis of pancreatic cancer, PLoS One 9 (1) (2014) [122] A. Chow, W. Zhou, L. Liu, M.Y. Fong, J. Champer, D. Van Haute, et al.,
e85942. Macrophage immunomodulation by breast cancer-derived exosomes
[96] J. Ellermeier, J. Wei, P. Duewell, S. Hoves, M.R. Stieg, T. Adunka, et al., requires Toll-like receptor 2-mediated activation of NF-kappaB, Sci. Rep. 4
Therapeutic efficacy of bifunctional siRNA combining TGF-beta1 silencing (2014) 5750.
with RIG-I activation in pancreatic cancer, Cancer Res. 73 (6) (2013) [123] M. Fabbri, A. Paone, F. Calore, R. Galli, E. Gaudio, R. Santhanam, et al.,
1709–1720. MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory
[97] A.T. Plegmasia alba dolens, Plegmasia alba dolens, Lect. Clin. Med. 18655 response, Proc. Natl. Acad. Sci. U. S. A. 109 (31) (2012) E2110–6.
(2016) 281–332. [124] S.A. Melo, L.B. Luecke, C. Kahlert, A.F. Fernandez, S.T. Gammon, J. Kaye, et al.,
[98] W.L. Dean, M.J. Lee, T.D. Cummins, D.J. Schultz, D.W. Powell, Proteomic and Glypican-1 identifies cancer exosomes and detects early pancreatic cancer,
functional characterisation of platelet microparticle size classes, Thromb. Nature 523 (7559) (2015) 177–182.
Haemost. 102 (4) (2009) 711–718. [125] R. Kalluri, The biology and function of exosomes in cancer, J. Clin. Invest. 126
[99] M. Dovizio, S. Alberti, A. Sacco, P. Guillem-Llobat, S. Schiavone, T.J. Maier, (4) (2016) 1208–1215.
et al., Novel insights into the regulation of cyclooxygenase-2 expression by [126] C. Zijlstra, W. Stoorvogel, Prostasomes as a source of diagnostic biomarkers
platelet-cancer cell cross-talk, Biochem. Soc. Trans. 43 (4) (2015) 707–714. for prostate cancer, J. Clin. Invest. 126 (4) (2016) 1144–1151.
[100] L. Erpenbeck, M.P. Schon, Deadly allies: the fatal interplay between platelets [127] J. Lu, G. Getz, E.A. Miska, E. Alvarez-Saavedra, J. Lamb, D. Peck, et al.,
and metastasizing cancer cells, Blood 115 (17) (2010) 3427–3436. MicroRNA expression profiles classify human cancers, Nature 435 (7043)
[101] L.J. Gay, B. Felding-Habermann, Contribution of platelets to tumour (2005) 834–838.
metastasis, Nat. Rev. Cancer 11 (2) (2011) 123–134. [128] R.J. Bryant, T. Pawlowski, J.W. Catto, G. Marsden, R.L. Vessella, B. Rhees, et al.,
[102] E. Sierko, M.Z. Wojtukiewicz, Inhibition of platelet function: does it offer a Changes in circulating microRNA levels associated with prostate cancer, Br.
chance of better cancer progression control? Semin. Thromb. Hemost. 33 (7) J. Cancer 106 (4) (2012) 768–774.
(2007) 712–721. [129] Z.H. Chen, G.L. Zhang, H.R. Li, J.D. Luo, Z.X. Li, G.M. Chen, et al., A panel of five
[103] S. Karpatkin, E. Pearlstein, C. Ambrogio, B.S. Coller, Role of adhesive proteins circulating microRNAs as potential biomarkers for prostate cancer, Prostate
in platelet tumor interaction in vitro and metastasis formation in vivo, J. 72 (13) (2012) 1443–1452.
Clin. Invest. 81 (4) (1988) 1012–1019. [130] J. Shen, G.W. Hruby, J.M. McKiernan, I. Gurvich, M.J. Lipsky, M.C. Benson,
[104] L. Borsig, R. Wong, J. Feramisco, D.R. Nadeau, N.M. Varki, A. Varki, Heparin et al., Dysregulation of circulating microRNAs and prediction of aggressive
and cancer revisited: mechanistic connections involving platelets, prostate cancer, Prostate 72 (13) (2012) 1469–1477.
P-selectin, carcinoma mucins, and tumor metastasis, Proc. Natl. Acad. Sci. U. [131] P.S. Mitchell, R.K. Parkin, E.M. Kroh, B.R. Fritz, S.K. Wyman, E.L.
S. A. 98 (6) (2001) 3352–3357. Pogosova-Agadjanyan, et al., Circulating microRNAs as stable blood-based
[105] B. Nieswandt, M. Hafner, B. Echtenacher, D.N. Mannel, Lysis of tumor cells markers for cancer detection, Proc. Natl. Acad. Sci. U. S. A. 105 (30) (2008)
by natural killer cells in mice is impeded by platelets, Cancer Res. 59 (6) 10513–10518.
(1999) 1295–1300. [132] N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki, T. Ochiya,
[106] G. Berchem, M.Z. Noman, M. Bosseler, J. Paggetti, S. Baconnais, E. Le Cam, Secretory mechanisms and intercellular transfer of microRNAs in living
et al., Hypoxic tumor-derived microvesicles negatively regulate NK cell cells, J. Biol. Chem. 285 (23) (2010) 17442–17452.
function by a mechanism involving TGF-beta and miR23a transfer, [133] L. Zitvogel, A. Regnault, A. Lozier, J. Wolfers, C. Flament, D. Tenza, et al.,
Oncoimmunology 5 (4) (2016) e1062968. Eradication of established murine tumors using a novel cell-free vaccine:
[107] L. Rong, R. Li, S. Li, R. Luo, Immunosuppression of breast cancer cells dendritic cell-derived exosomes, Nat. Med. 4 (5) (1998) 594–600.
mediated by transforming growth factor-beta in exosomes from cancer [134] J.A. Cho, Y.S. Lee, S.H. Kim, J.K. Ko, C.W. Kim, MHC independent anti-tumor
cells, Oncol. lett. 11 (1) (2016) 500–504. immune responses induced by Hsp70-enriched exosomes generate tumor
[108] M. Labelle, S. Begum, R.O. Hynes, Direct signaling between platelets and regression in murine models, Cancer Lett. 275 (2) (2009) 256–265.
cancer cells induces an epithelial-mesenchymal-like transition and [135] J. Wolfers, A. Lozier, G. Raposo, A. Regnault, C. Thery, C. Masurier, et al.,
promotes metastasis, Cancer Cell 20 (5) (2011) 576–590. Tumor-derived exosomes are a source of shared tumor rejection antigens
[109] E.U. Wieckowski, C. Visus, M. Szajnik, M.J. Szczepanski, W.J. Storkus, for CTL cross-priming, Nat. Med. 7 (3) (2001) 297–303.
Whiteside TL: tumor-derived microvesicles promote regulatory T cell [136] J.A. Cho, D.J. Yeo, H.Y. Son, H.W. Kim, D.S. Jung, J.K. Ko, et al., Exosomes: a
expansion and induce apoptosis in tumor-reactive activated CD8+ T new delivery system for tumor antigens in cancer immunotherapy, Int. J.
lymphocytes, J. Immunol. 183 (6) (2009) 3720–3730. Cancer 114 (4) (2005) 613–622.
[110] A.J. Abusamra, Z. Zhong, X. Zheng, M. Li, T.E. Ichim, J.L. Chin, et al., Tumor [137] C. Kahlert, R. Kalluri, Exosomes in tumor microenvironment influence
exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis, Blood Cells cancer progression and metastasis, J. Mol. Med. 91 (4) (2013) 431–437.
Mole. Dis. 35 (2) (2005) 169–173. [138] L. Elsner, V. Muppala, M. Gehrmann, J. Lozano, D. Malzahn, H. Bickeboller,
[111] T.L. Whiteside, Immune modulation of T-cell and NK (natural killer) cell et al., The heat shock protein HSP70 promotes mouse NK cell activity against
activities by TEXs (tumour-derived exosomes), Biochem. Soc. Trans. 41 (1) tumors that express inducible NKG2D ligands, J. Immunol. 179 (8) (2007)
(2013) 245–251. 5523–5533.
[112] M. Szajnik, M. Czystowska, M.J. Szczepanski, M. Mandapathil, T.L. Whiteside, [139] F. Andre, N.E. Schartz, M. Movassagh, C. Flament, P. Pautier, P. Morice, et al.,
Tumor-derived microvesicles induce, expand and up-regulate biological Malignant effusions and immunogenic tumour-derived exosomes, Lancet
activities of human regulatory T cells (Treg), PLoS One 5 (7) (2010) e11469. 360 (9329) (2002) 295–305.
[140] B. Escudier, T. Dorval, N. Chaput, F. Andre, M.P. Caby, S. Novault, et al.,
Vaccination of metastatic melanoma patients with autologous dendritic cell

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006
G Model
YSCBI-1296; No. of Pages 12 ARTICLE IN PRESS
12 T.B. Steinbichler et al. / Seminars in Cancer Biology xxx (2017) xxx–xxx

(DC) derived-exosomes: results of thefirst phase I clinical trial, J. Transl. [145] M.S. Kim, M.J. Haney, Y. Zhao, V. Mahajan, I. Deygen, N.L. Klyachko, et al.,
Med. 3 (1) (2005) 10. Development of exosome-encapsulated paclitaxel to overcome MDR in
[141] M.A. Morse, J. Garst, T. Osada, S. Khan, A. Hobeika, T.M. Clay, et al., A phase I cancer cells, Nanomed.: Nanotechnol. Biol. Med. 12 (3) (2016) 655–664.
study of dexosome immunotherapy in patients with advanced non-small [146] S.A. Kooijmans, P. Vader, S.M. van Dommelen, W.W. van Solinge, R.M.
cell lung cancer, J. Transl. Med. 3 (1) (2005) 9. Schiffelers, Exosome mimetics: a novel class of drug delivery systems, Int. J.
[142] A. de la Fuente, L. Alonso-Alconada, C. Costa, J. Cueva, T. Garcia-Caballero, R. Nanomed. 7 (2012) 1525–1541.
Lopez-Lopez, et al., M-Trap: exosome-based capture of tumor cells as a new [147] X. Zhang, Z. Pei, J. Chen, C. Ji, J. Xu, X. Zhang, et al., Exosomes for
technology in peritoneal metastasis, J. Natl. Cancer Inst. 107 (9) (2015). immunoregulation and therapeutic intervention in cancer, J. Cancer 7 (9)
[143] M. Airoldi, D. Amadori, S. Barni, S. Cinieri, S. De Placido, A. Di Leo, et al., (2016) 1081–1087.
Clinical activity and cardiac tolerability of non-pegylated liposomal [148] M. Trivedi, M. Talekar, P. Shah, Q. Ouyang, M. Amiji, Modification of tumor
doxorubicin in breast cancer: a synthetic review, Tumori 97 (6) (2011) cell exosome content by transfection with wt-p53 and microRNA-125b
690–692. expressing plasmid DNA and its effect on macrophage polarization,
[144] T. Boulikas, Clinical overview on Lipoplatin: a successful liposomal Oncogenesis 5 (8) (2016) e250.
formulation of cisplatin, Expert Opin. Investig. Drugs 18 (8) (2009)
1197–1218.

Please cite this article in press as: T.B. Steinbichler, et al., The role of exosomes in cancer metastasis, Semin Cancer Biol (2017),
http://dx.doi.org/10.1016/j.semcancer.2017.02.006

You might also like