Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

Cellular Oncology (2018) 41:223–252

https://doi.org/10.1007/s13402-018-0378-4

REVIEW

The versatile role of exosomes in cancer progression: diagnostic


and therapeutic implications
Vignesh Sundararajan 1 & Fazlul H. Sarkar 2 & Thamil Selvee Ramasamy 3,4

Accepted: 20 March 2018 / Published online: 17 April 2018


# International Society for Cellular Oncology 2018

Abstract
Background Recent advances in cancer biology have highlighted the relevance of exosomes and nanovesicles as
carriers of genetic and biological messages between cancer cells and their immediate and/or distant environments.
It has been found that these molecular cues may play significant roles in cancer progression and metastasis. Cancer
cells secrete exosomes containing diverse molecules that can be transferred to recipient cells and/or vice versa to
induce a plethora of biological processes, including angiogenesis, metastasis formation, therapeutic resistance,
epithelial-mesenchymal transition and epigenetic/stemness (re)programming. While exosomes interact with cells with-
in the tumour microenvironment to promote tumour growth, these vesicles can also facilitate the process of distant
metastasis by mediating the formation of pre-metastatic niches. Next to their tumour promoting effects, exosomes
have been found to serve as potential tools for cancer diagnosis and therapy. The ease of isolating exosomes and
their content from different body fluids has led to the identification of diagnostic and prognostic biomarker signa-
tures, as well as to predictive biomarker signatures for therapeutic responses. Exosomes can also be used as cargos
to deliver therapeutic anti-cancer drugs, and they can be engineered to serve as vaccines for immunotherapy.
Additionally, it has been found that inhibition of exosome secretion, and thus the transfer of oncogenic molecules,
holds promise for inhibiting tumour growth. Here we provide recent information on the diverse roles of exosomes in
various cellular and systemic processes governing cancer progression, and discuss novel strategies to halt this
progression using exosome-based targeted therapies and methods to inhibit exosome secretion and the transfer of
pro-tumorigenic molecules.
Conclusions This review highlights the important role of exosomes in cancer progression and its implications for (non-invasive)
diagnostics and the development of novel therapeutic strategies, as well as its current and future applications in clinical trials.

Keywords Exosomes . Extracellular vesicles . Metastasis . Tumour microenvironment . Anti-cancer therapeutics . Pre-metastatic
niche . Angiogenesis . Metastasis . Drug resistance . Epithelial-mesenchymal transition

1 Introduction
* Thamil Selvee Ramasamy
selvee@ummc.edu.my Cells use different methods to communicate with each other
1
and with their surrounding environment through direct contact
Department of Biotechnology, School of Bioengineering, SRM
Institute of Science and Technology, Kattankulathur, Tamil
or the secretion of soluble factors [1]. During cancer progres-
Nadu 603203, India sion, cancer cells communicate with neighbouring cancerous
2
Department of Pathology, Wayne State University School of
and non-cancerous cells, including mesenchymal stromal
Medicine, Detroit, MI 48201, USA cells, fibroblasts and endothelial cells, that collectively play
3
Stem Cell Biology Laboratory, Department of Molecular Medicine,
crucial roles in cancer development [2]. The three major types
Faculty of Medicine, University of Malaya, 50603 Kuala of communication that cells use effectively are: active trans-
Lumpur, Malaysia port, passive transport and vesicular transport. Of the modes
4
Cell and Molecular Biology Laboratory, Faculty of Medicine Dean’s of vesicular transport via exosomes, i.e., micro-vesicles and
Office, University of Malaya, 50603 Kuala Lumpur, Malaysia apoptotic bodies, the former has been intensely investigated in
224 V. Sundararajan et al.

recent years [1, 3]. Exosomes with a diameter of 50 nm to Unlike apoptotic bodies and other vesicles, the formation and
100 nm are secreted by a wide range of cell types [4], includ- release of exosomes follow distinct pathways. The first step in
ing cancer cells, to perform a myriad of functions, such as the the formation of exosomes involves inward budding of the
modulation of immune function [5], the regulation of cell membranes of endosomes to form MVEs with intraluminal
metabolism [6], conferring drug resistance, and promoting vesicles (ILVs). The second step involves the release of
metastasis and angiogenesis [7]. The presence of multi- exosomes. MVEs can either undergo degradation in lyso-
vesicular endocytic compartments determines whether cells somes or fuse with the plasma membrane to release the ILVs
are capable of mediating exosomal secretion [4]. Exosomes, as exosomes [4, 32]. Additional evidence strengthening the
which are clustered under extracellular vesicles (EVs), are endosomal origin of exosomes comes from the observation
secreted by almost all mammalian cell types, as well as by that the proteins found in the exosomes have cytosolic origins,
some lower eukaryotes and prokaryotes [1, 8, 9]. Cells may especially from endocytic compartments. Endocytic proteins
secrete exosomes in response to a range of stimuli, including that have been found in exosomes include annexin II, RAB5/
p53 activation [10, 11], ceramide synthesis [12], intracellular RAB7 and TSG101 [4]. An important component required for
calcium concentration alteration [13], pH changes [14], plas- the budding of endosomes into MVEs is the sphingolipid cer-
ma membrane depolarization [15, 16], T-cell receptor activa- amide [32]. Trajkovic et al. found that exosomes contain high
tion [17], hypoxia and ionising radiation [18]. How exosome amounts of ceramide and the enzyme sphingomyelinase
biogenesis is regulated and what their impact is on recipient (SMase), which facilitates the formation of ceramide. They
cells, which is largely determined by their content, will be also found that treatment of cells with a neutral SMase inhib-
discussed in the next section. itor significantly inhibited exosome biogenesis and subse-
quent release [32], suggesting an important role of this en-
zyme in exosome biogenesis, which will be discussed in the
2 Exosome structure, biogenesis and content next section.

The term ‘exosome’ was first coined by Tram et al. in 1981 as it 2.2 Exosome biogenesis
was found that cultured normal and neoplastic cells may secrete
vesicles with 5′-nucleotidase activity [19]. The process of The formation of MVEs can occur either in an Endosomal
exosome secretion was subsequently described in further detail Sorting Complex Required for Transport (ESCRT)-dependent
by Pan et al. and Harding et al. after studying endocytosis and or an ESCRT-independent manner. The ESCRT machinery is
the intracellular processing of transferrin receptors (TR) in re- composed of the ESRT-0, -I, -II and -III complexes. These
ticulocytes. It was found that colloidal gold-TF was incorporat- complexes sort ubiquitinated intracellular cargos, such as
ed into multi-vesicular endosomes (MVEs) as well as inclusion internalised receptors, into MVEs and, by doing so, prevent
vesicles, and that these inclusion vesicles contained TR- their degradation in lysosomes [33]. The lysosomal/late
receptors that were shed from developing reticulocytes via exo- endosomal degradation of the epidermal growth factor recep-
cytosis [20, 21]. Exosomes may contain various biomolecules tor (EGFR) is, for example, catalysed by ubiquitination of the
such as mRNAs, microRNAs (miRNAs), long non-coding EGFR upon its exit from early endosomes to lysosome/late
RNAs (lncRNAs), DNA and proteins [22]. Intriguingly, it has endosomes by the E3 ligase c-Cbl [34, 35]. The release of
been found that the contents of exosomes from donor cells can MVEs from a cell begins with the binding of ESCRT-0 to
be transferred to recipient cells and that the DNA/RNA and endosomes and its subsequent interaction with the ubiquitin
protein molecules derived from these exosomes can function- moieties of the cargos that would normally undergo lysosomal
ally modulate cellular processes within recipient cells [23]. This degradation [36]. Whereas ESCRT-0 forms domains of clus-
type of communication may involve a wide range of cell types, tered cargos, ESCRT-I and ESCRT-II facilitate the formation
including normal and tumour cells, fibroblasts and endothelial of buds in the membranes of endosomes. The ESCRT-0-
cells. Currently, exosomes have been isolated and purified from ubiquitin domains are recruited to the buds, followed by
various sources of body fluids, such as breast milk [24], blood ESCRT-III which cleaves the buds to form ILVs [33].
plasma [25], cerebrospinal fluid [26], saliva [27], peritoneum Although ESCRT is considered to be important for the forma-
lavage fluid [28], urine [29] and serum [30], which underscores tion of ILVs, it has been found that ILV synthesis may also
the putative importance of exosomes and their roles as messen- occur after ESCRT inhibition. The components of the
gers between cells, tissues and organs. endocytic pathway were found to exhibit morphological
changes in ESCRT-depleted cells, but clearly maintained dif-
2.1 Exosome structure ferences between early and late endosomes [37]. Important
components of the ESCRT-independent pathway include ac-
Exosomes are cup-shaped vesicles with a lipid bilayer that is tivated sphingosine 1-phosphate [38] and the tetraspanin-
enriched in cholesterol, sphingomyelin and ceramide [12, 31]. enriched microdomain (TEM) interactome [39].
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 225

2.3 Exosome content further in-depth investigation on how exosomes can be tailored
to express ligands on their surface corresponding to receptors that
Since cancer cells communicate with each other and other cell are (over)expressed by cancer cells.
types via exosomes through the messages that they carry, the
exosome content must be variable and specific in order to
convey different messages. The exosome database (www. 3 Role of Rab GTPases and other regulators
exocarta.org) lists 9769 proteins, 3408 mRNAs and 2838 in exosome secretion
miRNAs in its latest update, several of which play important
roles in tumour progression and metastasis through the Dysregulation of Rab GTPases has been encountered in sev-
creation of a vicious tumour microenvironment. Some of the eral cancers [52]. This family of enzymes is also known for its
most commonly found proteins in exosomes, which reflect role in exosome secretion and transport, as well as in mem-
their endosomal origin, include membrane transport and brane trafficking. Different family members are known to reg-
fusion proteins (GTPases, annexins, flotillin), tetraspanins ulate the secretion of small- and/or larger-sized exosomes and
(CD9, CD63, CD81, CD82), heat shock proteins (Hsp70, the transport of specific biomolecules, including miRNAs. It
Hsp90), multi-vesicular body synthesis proteins (Alix, has been found, for example, that the transport of miR-143 by
TSG101) and lipid related proteins [40]. Exosomes may also exosomes from endothelial cells, induced by the shear stress
transport different lipids such as sphingolipids, cholesterol responsive transcription factor Krüppel-like factor 2 (KLF2),
and ceramide, which are components of lipid rafts. They have depends on the expression of Rab7a and Rab27b.
also been reported to transport signalling molecules such as Interestingly, it was also found that the KLF2-induced
phospholipase A2 (PLA2), arachidonic acid and prostaglan- exosomal export of miR-143 was not dependent on Rab27a,
dins [7, 41]. Some of the lipids secreted by exosomes, such as a homologue of Rab27b, which is in accordance with the
lysophosphatidylcholine and PLA2, are known to modulate notion that Rab family GTPases exhibit cell type-specific
immune responses by favouring dendritic cell (DC) matura- functions [53]. Whereas it has been found that Rab5B,
tion [42, 43]. In addition, exosomes are known to contain Rab9A and Rab17 expression is associated with the produc-
nucleotides such as mRNAs, lncRNAs and miRNAs. Highly tion of small-sized exosomal vesicles, Rab32 has been found
stable RNAse resistant nucleic acids, especially miRNAs [44], to mediate the secretion of larger-sized exosomal vesicles
can be transferred to recipient cells to confer oncogenic and [54]. It has also been found that inhibition of Rab35 may lead
non-oncogenic functions, which will be discussed in further to a reduction of the mean size of the readily releasable pool
detail in Section 4.2.2. Internalization of exosomal contents in (RRP) of exomes. Rab35 is a target of TBC1D10A-C, and its
recipient cells may occur via their fusion with target cell mem- inhibition has been found to lead to impairment of exosome
branes, clathrin-mediated endocytosis, caveolin-mediated en- secretion [55].
docytosis, lipid raft-mediated endocytosis, macro-pinocytosis, It has been reported that the activities of different Rab
receptor-ligand interactions and/or phagocytosis [45–47]. GTPases may be regulated by different factors, including in-
There is ambiguity regarding the question whether any cell tracellular Ca2+ concentration [35], hypoxia-inducible factor-
can take up exosomes or whether this process is cell-type specif- 1α (HIF-1α) expression [37], heparanase expression [56] and/
ic. It has, for example, been reported that exosomes secreted by or microenvironmental pH changes [14]. The release of
glioblastoma cells can be taken up by various normal and trans- exosomes from the RRP mediated by Rab11 and Rab35 has,
formed cells [48]. Although exosomes secreted by rat pancreatic for example, been found to be stimulated by intracellular Ca2+
adenocarcinoma cells were found to be taken up in vitro and [55]. Rab11 plays an important role in the docking and fusion
in vivo by a variety of leukocytes, this uptake was found to be of multi-vesicular bodies (MVBs), but the release of exosomes
associated with the presence of ligands on leukocytes that could has been observed only in the presence of increased intracel-
interact with exosomal receptors [49]. Also, others reported a lular Ca2+ concentrations [57]. The release of exosomes may
cell-type specific uptake of exosomes. Rana et al., for example, also be accelerated under hypoxic conditions, and this effect
found that exosomes secreted by normal lymph node stromal has been found to be mediated by HIF-1α [58]. It has been
cells expressing the Tspan8-alpha4 complex were most readily reported that under hypoxic conditions tumour masses may
taken up by endothelial and pancreatic cells [50]. It has also been undergo several changes, including the formation of cancer
reported that cell-type specific uptake of exosomes may require stem cells (CSCs), increased angiogenesis and the activation
the presence of ligands, such as MUC1, on their surface, which of cancer survival mechanisms [59]. Hence, cancer cells need
can interact with specific receptors, such as the dendritic cell- to modulate their microenvironment in order to facilitate their
specific intercellular adhesion molecule-3 grabbing non-integrin growth and further transformation. This modulation may in-
(DC-SIGN), present on the surface of target cells [51]. The cell- clude the recruitment of cancer-associated cells and (excreted)
type specific uptake of exosomes may be employed for the de- stimulatory factors. These reciprocal interactions have been
sign of novel targeted therapeutic approaches and warrants shown to be mediated by exosomes. Hence, the targeting of
226 V. Sundararajan et al.

exosome-based communication mechanisms may serve as a Fig. 1 Schematic representation of exosomal molecules secreted by„ b
therapeutic strategy. The validity of this notion has already cancer cells and cells of the tumour microenvironment that drive
different events leading to therapy failure, distant metastasis and the
been shown in some clinical trials, which will be discussed
acquisition of aggressive phenotypes. Inhibition of exosomal secretion
below. may represent a novel targeted strategy to treat drug-resistant tumours
as well as those with a high metastatic capacity

4 Role of exosomes in cancer progression cytoplasmic and nuclear β-catenin levels might be responsible
for the observed increased motility, invasion and stemness of
Exosomes secreted by both tumour cells and tumour- these cells [63]. It has also been shown that tumour cells may
associated cells play predominant roles in favouring tumour adopt dormancy or the acquisition of a quiescent state at met-
growth via the transfer of pro-tumorigenic factors (Fig. 1). astatic sites for certain periods of time until cues for tumour re-
Glioma cells, for example, may secrete exosomes containing growth and progression become available. Exosomes released
EGFRvIII that can promote the growth of recipient cells that by cancer cells may promote tumour growth and metastasis at
lack this truncated epidermal growth factor receptor variant distant sites. They may also determine the site of metastasis
through the activation of transforming signalling pathways, based on the presence of specific factors, such as integrins, on
such as the Akt and mitogen-activated protein kinase their surface. In the following sections different mechanisms
(MAPK) pathways. In addition, it has been found that of exosome-mediated cancer progression will be discussed.
exosomes containing EGFRvIII may promote angiogenesis
by enhancing the expression of the vascular endothelial
growth factor (VEGF), as well as cellular proliferation and 4.1 Role of exosomes in cancer growth
survival by increasing the expression of the anti-apoptotic
protein Bcl-xL and by downregulating the expression of 4.1.1 Angiogenesis
p27, a cyclin-dependent kinase (CDK) inhibitor [60]. The
presence of inhibitors of apoptosis (IAP), such as survivin, Tumours require a continuous nutrient and oxygen supply
XIAP, cIAP1 and cIAP2, in exosomes secreted from cancer through blood circulation. In a hypoxic milieu CSCs favour
cells may also protect cancer cells from apoptosis and favour angiogenesis via the secretion of VEGF, which promotes en-
tumour growth [61]. More recently, it has been found that dothelial cell migration and tube formation [64]. In addition,
exosomes may confer aggressive phenotypes to cancer cells CSCs may contribute to the growth of new blood vessels by
through stemness regulators such as miRNAs. Breast cancer differentiating into endothelial cells [65] and by secreting pro-
cells treated with exosomes secreted by cancer-associated fi- angiogenic factors such as angiopoietin [66]. It has also been
broblasts (CAFs) containing miR-21, -378e and -143 were, for reported that exosomes secreted by both tumour cells and
example, found to exhibit increased anchorage independent CSCs may promote angiogenesis, but that these exosomes
growth and mammosphere forming abilities concomitant with differ significantly in their content and their ability to promote
upregulation of the expression of stem cell markers (Oct3/4, tumour growth. It has been found, for example, that exosomes
Nanog, Sox2) and epithelial-mesenchymal transition (EMT) secreted by CD105+ CSCs contain high levels of 24 miRNAs
regulators (Snail and Zeb) [62]. These results suggest a role of regulating transcription, metabolic processing, proliferation,
the tumour microenvironment in modulating cancer cell fate nucleic acid binding and cell adhesion molecules [67]. In ad-
and aggressiveness, resulting in tumour metastasis and re- dition, the authors found that only exosomes secreted by
lapse. Similarly, exosomes secreted by LNCaP and PC3 pros- CD105+ CSCs contained pro-angiogeneic mRNAs such as
tate cancer cells under hypoxic conditions have been reported those encoding VEGF, fibroblast growth factor (FGF),
to increase the stemness of naïve LNCaP and PC3 cells. When angiopoietin1, ephrin A3, MMP-2, and MMP-9 and growth
cultured in the presence of exosomes, significant increases in factors. CD105+ CSC-derived exosomes were also found to
prostasphere formation were observed in these cells. This was be more effective than exosomes derived from CD105− tumour
due to the phenotypic conversion of fibroblasts into CAFs. cells in favouring angiogenesis. Others found that CD90+ liver
Importantly, it was found that exosomes derived from hypoxic CSCs secrete exosomes containing lncRNA H19, which may
prostate cancer cells may contain more proteins such as stimulate angiogenesis by favouring tube formation and en-
tetraspanins, HSP and annexin II, matrix metalloproteinases hancing the adhesions between CD90+ cells and endothelial
(MMPs) such as MMP-2 and MMP-9, cytokines and signal- cells by upregulating the expression of intercellular adhesion
ling molecules such as TGF-β2, tumour necrosis molecule 1 (ICAM-1) [68]. Since only a limited number of
factor-(TNF)-1α, interleukin (IL) 6 (IL6), Akt, integrin- studies has been published on the occurrence of differences in
linked kinase 1 (ILK1), and β-catenin, than those derived composition and function of exosomes secreted by CSCs and
from normoxic cells, and the authors postulated that loss of tumour cells, and the notion that CSC-derived exosomes may
E-cadherin expression and concomitant increases in be more tumorigenic, additional studies are warranted.
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 227
228 V. Sundararajan et al.

Nevertheless, it can be postulated that it may be promising to been found that exosomes secreted by leukaemia cells under
develop therapeutic strategies aimed at both inhibiting exosome hypoxic conditions may contain elevated levels of miR-210,
secretion by CSCs and targeted elimination of CSCs. and that these exosomes enhance tube formation in HUVECs
WNT5A signalling in melanoma cells may trigger Ca2+- by eliciting expression downregulation of Ephrin A3
dependent release of exosomes, and it has been found that (EFNA3), an anti-angiogenic factor [74]. Similarly, multiple
these exosomes contain immunomodulatory and pro- myeloma cells under hypoxic conditions have been found to
angiogenic factors, such as IL6, VEGF and MMP-2. These secrete exosomes containing high levels of miR-135b, which
factors, in turn, may enhance the immunosuppressive capacity favours tube formation in endothelial cells through inhibition
of tumours as well as endothelial cell branching to confer of HIF-1α [75]. Thus, exosomes secreted by tumour cells may
more aggressive and metastatic phenotypes [69]. A recent secrete pro-angiogenic factors and miRNAs that modulate the
study has shown that cancer cells undergoing EMT may se- phenotype of endothelial cells to promote angiogenesis.
crete exosomes containing Rac1/p21-activated kinase-2
(PAK-2) and, by doing so, communicate with endothelial cells 4.1.2 Epithelial-mesenchymal transition
to promote angiogenesis [70]. Co-treatment of endothelial 2F-
2B cells with exosomes secreted by tumour cells undergoing A number of processes, such as the transition of epithelial cells
EMT increased the motility of these cells, enhanced the num- to mesenchymal cells, extracellular matrix (ECM) remodel-
ber of vessels formed and increased their length, again indi- ling, the acquisition of unique molecular signatures (including
cating that exosome-mediated communication is imperative stemness and resistance markers) and cytoskeletal reorganiza-
for cancer progression. Conversely, it was found that inhibi- tion take place during EMT. A classical hallmark of EMT is a
tion of downstream targets of Rac1, such as PAK-2, reduced decrease in E-cadherin expression and a concomitant increase
tube formation, tube length and branch points of endothelial in the expression of several transcription factors, such as Snail,
cells, indicating that exosomal Rac1 signalling in endothelial Slug and Twist, and N-cadherin [76]. It has also been reported
cells may serve as a therapeutic target. Cigarette smoke extract that the process of EMT requires support from tumour-
(CSE)-transformed human bronchial epithelial (HBE) cells associated cells. The treatment of nasopharyngeal carcinoma
have been found to secrete signal inducer and activator of (NPC) cells with exosomes secreted by mesenchymal stem/
transcription-3 (STAT-3)-regulated exosomal miR-21, which stromal cells (MSCs) enriched in FGF19 isolated from bone
can be transferred to normal HBE cells to change their phe- marrow of healthy donors has, for example, been found to
notype towards a transformed state and, subsequently, in- result in EMT-like changes in cellular characteristics such as
crease their VEGF expression to promote angiogenesis. an elongated morphology and an increased expression of the
Additionally, it was found that treatment of human umbilical mesenchymal markers N-cadherin and vimentin, thereby con-
vein endothelial cells (HUVECs) with exosomes derived from ferring increased migratory abilities to these cells.
CSE-transformed HBE cells containing miR-21 increased Mechanistically, FGF19 increased the expression of pERK
their tube formation, and that their relative tube length was and phosphorylated fibroblast growth factor
increased in a dose-dependent manner [71]. The control over receptor-(FGFR)-4 (pFGFR4), which subsequently modulat-
angiogenesis by switching to higher pro-angiogenic and ed the expression of EMT markers [77]. Knowing that MSCs
lower anti-angiogenic protein levels has been underscored secrete factors that favour tissue regeneration and repair via
by studies in which exosomes derived from nasopharyn- the induction of proliferation, migration and survival signals,
geal cancer cells were investigated. These exosomes were and by suppressing apoptosis, it seems plausible to assume
found to contain elevated levels of pro-angiogenic pro- that those same exosomes may accelerate tumour growth, a
teins, such as ICAM-1 and CD44v5, and decreased levels notion that should be taken into account in future studies.
of the anti-angiogenic protein thrombospondin-1 (TSP-1). Moreover, as discussed below, there are studies suggesting a
Internalization of these exosomes by HUVECs altered the role of MSCs in inhibiting the growth of cancer cells. This
levels of angiogenic proteins in these cells and, subse- raises the question whether MSCs may play differential roles
quently, elicited increased tube formation, migration and depending on the niche in which they reside and the presence
invasion [72]. Taken together, it appears that cancer cells of local environmental cues. Further in-depth studies aimed at
may secrete several factors through exosomes that can understanding the positive and negative roles of MSCs in
modulate a variety of their own cellular activities and tumour progression may provide novel leads to the develop-
those of their surrounding cells, thereby harnessing ment of effective therapeutic strategies.
tumour-associated cellular activities supporting tumour The phenomena of exosome-mediated increases in migra-
progression. tion and invasion have been observed in many other studies,
Additional lines of research have shown that hypoxia may including those dealing with the effects of exosomes secreted
favour angiogenesis by promoting the secretion of exosomes by bladder cancer cells on urothelial cells. It was found that
containing pro-angiogenic factors [73]. It has, for example, these urothelial cells exhibited enhanced invasive and migrative
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 229

capacities, which could subsequently be inhibited by blocking inoculation of the exosomes in mice led to an enhanced tu-
exosome uptake via heparin pre-treatment [78]. Epstein-Barr mour growth and an increased metastatic potential [84].
virus (EBV)-positive NPC cells contain increased levels of Exosomes derived from gastric cancer cells and malignant
HIF-1α in their secreted exosomes. The secretion of HIF-1α pleural effusions have been found to promote peritoneal me-
into the exosomes of these cells was augmented by the principal tastasis of gastric cancer cells by up-regulating the expression
oncoprotein of EBV, latent membrane protein 1 (LMP1). HIF- of adhesion molecules in mesothelial cells and, concomitantly,
1α secreted into these exosomes was found to be stable, as it increasing the migratory ability of gastric cancer cells [85].
retained its DNA-binding ability and transcriptional activity. These studies underscore the importance of tumour-derived
The uptake of these exosomes by EBV-negative NPC cells exosomes in the metastasis of tumour cells and provide new
was found to result in an increased migration and invasion directions for the treatment of metastases.
resulting from decreased E-cadherin and increased N-cadherin It has amply been shown that exosomal miR-21 performs
expression [79]. Intriguingly, exosomes can also be secreted diverse tumour-related functions, as discussed in various sec-
into human breast milk, and it has been reported that exosomes tions in this review. Specifically, it has been found that miR-21
from benign as well as malignant breast tumours contain high secreted via exosomes may promote the invasion and metas-
levels of transforming growth factor beta (TGF-β), a key factor tasis of cancer cells. Under hypoxic conditions, oral squamous
involved in EMT induction, which may upregulate the expres- cell carcinoma (OSCC) cells secrete exosomes containing el-
sion of the mesenchymal markers α-smooth muscle actin (α- evated miR-21 levels, and these exosomes have been found to
SMA) and vimentin, and downregulate the expression of E- increase the migration and invasion of normoxic OSCC cells
cadherin [80]. in a HIF-1α and HIF-2α-dependent manner [86]. It has also
It has also been found that miRNAs may play important been found that exosomes secreted by oesophageal cancer
roles in the process of EMT through modulating the expres- cells containing miR-21 can be taken up by oesophageal cells
sion of different EMT markers [81]. Primary melanocytes in a nSMase2-dependent manner [87]. Subsequent co-culture
may acquire metastatic capabilities and become aggressive experiments showed that miR-21 shuttling into recipient oe-
as a result of communication between melanoma cell-derived sophageal cancer cells increased their invasive and migrative
exosomes and primary melanocytes, and it has been found capacities. Mechanistically, miR-21 was found to downregu-
that the change towards a EMT-like phenotype results from late the expression of its target gene programmed cell death 4
activation of the MAPK pathway and a decreased expression (PCD4), and to activate c-Jun N-terminal kinase (JNK) down-
of miRNA let-7i. Exogenous re-expression of let-7i in primary stream signalling, resulting in enhanced expression of MMP-2
melanocytes using a let-7i mimic and its subsequent co- and MMP-9. Thus, exosomal miR-21 may serve as a thera-
culture with exosomes derived from melanoma cells was peutic exosomal target that warrants further investigation.
found to result in modulation of the expression of the let-7i The importance of interactions between macrophages and
targets LIN28B and HMGA2, and a concomitant decrease in tumour cells during metastasis development has been investi-
the migration and invasion of primary melanocytes due to E- gated by Yang et al. They reported the transfer of exosomal
cadherin up-regulation and vimentin down-regulation [82]. miR-223 from IL4 activated macrophages to co-cultured
Thus, these studies underscore the role of exosomes in en- breast cancer cells. In addition, they reported that transfection
hancing the invasive and metastatic capacities of tumour cells of a miR-223 mimetic into breast cancer cells increased their
through EMT, and provide additional indications for therapeu- invasiveness through modulation of the myocyte enhancer
tically targeting exosome uptake in recipient cells to reverse factor 2C (Mef2c)-β-catenin pathway. After miR-223 mimetic
EMT. transfection a decreased Mef2C expression was observed
which, in turn, favoured nuclear translocation of β-catenin
4.1.3 Metastasis resulting in increased breast cancer cell invasion [88].
Mef2C is a co-transcription factor that plays an important role
The treatment of metastasised tumours is a major clinical chal- in both normal cell differentiation and in cancer development
lenge, which may at least in part be attributed to vast hetero- [89]. In hepatocellular carcinoma (HCC) cells, Mef2C has
geneities in both tumour cell populations and their environ- been found to act either as a tumour suppressor or as a tumour
mental tissue niches [83]. It has amply been shown that cancer enhancer, depending on its subcellular localisation and its in-
cell derived-exosomes may play a role in the metastasis of teraction with either VEGF or β-catenin. A nuclear sub-
tumour cells by enhancing their migratory ability. Breast can- localisation of Mef2C favours angiogenesis via its interaction
cer cells that overexpress CXCR4 exhibit, for example, stem with VEGF, whereas a cytosolic sub-localisation suppresses
cell-like/migratory inducing properties, and exosomes secret- tumour growth by inhibiting the nuclear translocation of β-
ed by these cells have been found to be internalised by T47D catenin. In order to confirm the tumour inhibitory activity of
breast cancer cells. This internalisation led, subsequently, to Mef2C, its expression was upregulated in in vitro and in vivo
increased cancer cell invasion and migration in vitro, whereas mouse HCC xenograft models. By doing so, it was indeed
230 V. Sundararajan et al.

found that Mef2C interacts with β-catenin and inhibits its and, finally, activation of the downstream Raf/MEK/ERK kinase
nuclear translocation, resulting in decreased cell proliferation cascade [96]. Since CSCs exhibit resistance to chemotherapeutic
in vitro and reduced tumour growth in vivo [89]. pressure, priming of CSCs with CAF-secreted exosomes may
Another recently discovered mechanism of metastasis pro- further increase drug resistance. Hence, targeting the secretion
motion by breast cancer cell-secreted exosomes has been re- of exosomes from both CSCs and CAFs may provide new ave-
ported by Fong et al. They found that exosome-secreted miR- nues to overcome drug resistance [95].
122 led to decreases in glucose uptake by non-tumour cells, Targeting cancer cells using antibodies directed against an-
such as lung fibroblasts, brain astrocytes and neurons, in pre- tigens present on their surface forms the basis of immunother-
metastatic niches resulting in enhanced metastasis. By decreas- apy, and acquired resistance to such therapies has delineated a
ing the expression of the glucose transporter GLUT1 and that of role of exosomes in this resistance. It has been found, for
pyruvate kinase M (PKM), an enzyme involved in glucose example, that B-cell lymphoma-secreted exosomes containing
metabolism, nutrient availability was increased facilitating the CD20 in an ATP-binding cassette (ABC) transporter A3
colonization of circulating cancer cells in lung and brain [90]. (ABCA3)-dependent manner can protect lymphoma cells
from antibody attack by binding anti-CD20 antibodies [97].
4.1.4 Drug resistance Similarly, it has been found that the effect of Trastuzumab can
be negated by human epidermal growth factor receptor 2
The development of resistance to chemotherapeutic drugs is one (HER2) secreted in exosomes from HER2-overexpressing
of the most common causes of treatment failure. The underlying breast cancer cells [98]. Drug-resistant tumour cells can also
molecular mechanisms include decreased uptake of anti-cancer reprogram lysosomal trafficking and secretion to enhance
drugs, increased expression of efflux transporters, enhanced exosome-mediated drug effluxes. Drug-resistant ovarian can-
DNA repair and detoxification of drugs [91]. These intrinsic cer cells may, for example, abnormally sort lysosomal proteins
mechanisms are linked to extrinsic mechanisms favouring the and secrete exosomes containing more cisplatin than drug
survival of cancer cells. These extrinsic mechanisms are repre- sensitive cells. Such cells have been found to exhibit a signif-
sented by, at least in part, exosome-based signalling. The major icant upregulation of genes associated with membrane fusion,
mechanisms involved in exosome mediated drug resistance in vesicle trafficking and the cisplatin efflux transporters MRP2,
cancer cells include (i) conferring resistant phenotypes to drug ATP7A and ATP7B [99].
sensitive cancer cells and (ii) recruiting tumour-associated cells Another mechanism by which exosomes may confer drug
that regulate protective mechanisms in cancer cells. It has been resistance to cancer cells is by secreting lncRNAs that can elicit
reported that transfer of miR-100, miR-222 and miR-30a from epigenetic changes. LincRNA-ROR (linc-ROR) is a long non-
drug resistant breast cancer cells to drug sensitive breast cancer coding RNA that was first described in 2010 for its role in the
cells can modulate drug-induced apoptosis and confer chemo- induction of pluripotent stem cells [100]. Subsequently, it was
resistance. Specifically, it was found that exosome-borne miR- found that linc-ROR expression is associated with a poor prog-
222 of docetaxel-resistant breast cancer cells can downregulate nosis in breast, pancreas, liver, endometrium, colon and naso-
the expression of its target gene phosphatase and tensin homolog pharyngeal cancer patients as it modulates the expression of
(PTEN) in recipient cells [92]. Another mechanism of conferring target genes governing tumour progression-associated mecha-
drug resistance to drug sensitive breast cancer cells involves nisms such as proliferation, invasion, metastasis and apoptosis
exosomal transfer of the efflux pump P-glycoprotein (P-gp) [101]. Takahashi et al. reported that TGF-β can mediate drug
[93]. Others have found that exosomes secreted by cancer- resistance in sorafenib-treated HCC cells by increasing linc-
associated adipocytes (CAAs) and CAFs in ovarian cancer pa- ROR levels in exosomes secreted by these cells. This, in turn,
tients may exhibit elevated miR-21 levels which, when trans- may repress p53 to inhibit apoptosis and enhance tumorigenic
ferred to ovarian cancer cells, may inhibit their apoptosis and capacities of recipient cells, including the induction of increases
confer chemo-resistance through downregulating the expression in tumour-initiating cells expressing CD133 and their ability to
of its target APAF1 [94]. Hu et al. reported yet another mecha- form colonies [102]. Although no linc-ROR elicited epigenetic
nism that may underlie chemo-resistance in a colon cancer model, changes were reported in this study, in another study by Fan
wherein exosomes secreted by CAFs primed CSCs to promote et al. it was reported that linc-ROR can promote tumour growth
their clonogenicity and growth when treated with anti-cancer and metastasis by acting as a decoy oncoRNA to inhibit meth-
drugs such as 5-flurouracil (5-FU) or oxaliplatin [95]. Gastric ylation of the TESC gene promoter through histone G9A meth-
cancer cells have been found to exhibit enhanced resistance to yltransferase and promoting the release of histone H3K9 meth-
5-FU through upregulation of multi-drug resistance (MDR) pro- ylation [103]. So, chemo-resistance in HCC cells may be due to
teins and activation of the CaM-Ks/Raf/MEK/ERK pathway via epigenetic modifications induced by exosome-secreted linc-
exosomes secreted by MSCs containing P-gp/MDR. Mechanistic ROR. Taken together, these studies highlight multiple mecha-
studies revealed that this drug resistance was due to influx of Ca2+ nisms involving exosomes in conferring drug resistance. This
via P-gp, followed by activation of calcium/calmodulin kinases information may be instrumental for the development of novel
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 231

therapeutic approaches aimed at sensitising drug-resistant cells senescence [111]. Interestingly, it has been found that both
to chemo- and/or immunotherapy. senescence-induced cellular aging and cancer are regulated
through multiple common regulators such as sirtuins and
4.1.5 Quiescence and senescence p53 [112], in which both these factors are known to play a
role in regulating exosome secretion.
As a result of a high therapeutic pressure and an unfavourable It has also been shown that senescence induction due to
environment, cancer cells may adopt multiple survival modes, Hayflick factors may result in an increased secretion of
including drug efflux and the acquisition of a quiescent or exosomes in a p53-dependent manner via upregulation of tar-
dormant state. The latter mechanism refers to a reversible state get genes such as the tumour suppressor-activated pathway 6
by which cancer cells arrest in the G0 phase of the cell cycle (TSAP6), CHMP4C and hepatocyte growth factor-regulated
with the ability to re-enter the cell cycle in the presence of tyrosine kinase substrate (HGS) genes [10, 113, 114]. The
favourable external stimuli. Cells can be maintained in this increased secretion of exosomes by senescent cells is referred
state for a long time through the regulation of genes associated to as senescence-associated secretome phenotype (SASP).
with cell cycle progression, DNA replication, mitochondrial Senescent cells secrete, via exosomes, various factors such
function, transcription regulation and RNA processing [104]. as growth factors, cytokines and extracellular proteases, which
When the tumour environment becomes favourable again, may act as signals to modulate the functions of neighbouring
these genes may be activated to cause tumour growth, relapse cells in the tumour environment [115]. In addition, senescent
and metastasis. A key problem is the variability of the dor- cells may release exosomes as a means to communicate their
mancy periods and, thus, the unpredictable occurrence of re- state and function with neighbouring cells and to favour tu-
lapses. Another problem is the usually more aggressive nature mour progression. It has, for example, been found that senes-
of the recurrences. The regulation of dormancy and quies- cent fibroblasts may secrete increased levels of VEGF to cause
cence is multifactorial and may, at least partially, be exosome in vivo tumour vascularisation in mice and to promote in vitro
mediated. It has, for example, been reported that dormant basement membrane invasion by HUVECs [116]. TSAP6 is
breast cancer cells in a bone environment may interact with an essential component of the p53-dependent exosome secre-
MSCs, causing the release of exosomes containing miR-222/ tion pathway as TSAP6−/− mice are unable to secrete
223. These exosomes lead to entry of the breast cancer cells exosomes following p53 activation [11]. In addition, it has
into a quiescent state by downregulating the expression of the been found that exosomes secreted by p53 deficient cancer
cell cycle regulatory proteins CDK4, cyclin D1 and p21WAF1. cells are smaller in size due to downregulation of its target
This downregulation may lead to a cell cycle arrest and, sub- gene HGS, compared to those secreted by wild-type p53 can-
sequently, a reduction in sensitivity to chemotherapy. cer cells [114].
Similarly, it has been found that exosomal transport of, Irradiation of prostate cancer cells with clinically relevant
amongst others, miR-127, miR-197, miR-222 and miR-223 doses may lead to premature senescence accompanied by an
by bone marrow stroma cells to breast cancer cells, and enhanced secretion of exosomes containing B7-H3, a protein
miR-23b from bone marrow MSCs to metastatic breast cancer that elicits anti-tumour immunity in a p53-dependent manner
cells, may induce breast cancer dormancy within the bone [117]. As such, detecting the levels of this protein may help to
[105, 106]. Interestingly, it has been reported that targeting predict responses to radiotherapy [118]. In a recent study, the
these dormant breast cancer cells with antagomiR-222/223 development of resistance to paclitaxel in ovarian cancer cells
may re-sensitise them to chemotherapy [107]. Similar ap- was attributed to senescence induction by exosomal miR-433
proaches may be relevant for therapeutically targeting other secreted by high miR-433 expressing cancer cells. These se-
dormant cancer cells. nescent ovarian cancer cells were found to exhibit altered cell
In contrast to quiescence, senescence refers to a presum- cycle states and to secrete significantly higher levels of the pro-
ably irreversible arrest of cells in response to Hayflick factors, inflammatory cytokines IL6 and IL8. Cell cycle progression in
i.e., telomere shortening, transcriptional depression of the ovarian cancer cells with increased miR-433 expression was
INK4a/ARF locus and DNA damage. Senescence is used as affected due to decreased hyperphosphorylation of Rb
a protective mechanism to limit cell proliferation, usually due resulting from decreased CDK6 expression, a cyclin-
to aging and cellular stress, and to limit tumour growth [108]. dependent kinase involved in Rb phosphorylation [119]. It
Senescence induction facilitates cancer treatment as senescent has also been reported that miR-433-mediated decreases in
cells become arrested in the G1 phase of the cell cycle and cell mitotic arrest deficiency protein 2 (MAD2) levels may contrib-
proliferation is inhibited in response to anti-cancer drugs and/ ute to resistance to paclitaxel in patients with high-grade serous
or radiation [109]. Treatment failure in head and neck cancer epithelial ovarian cancer [120]. Consistent with these results,
has, for example, been associated with inhibition of radiation- the expression of two miR-433 target genes, HDAC6 and
induced senescence due to mutations in the TP53 gene [110], MAD2, was found to be decreased in cancer cells overexpress-
as also in invasive colon cancer through escape from ing miR-433, which may be responsible for the development
232 V. Sundararajan et al.

of drug resistance in these cells [119]. Conversely, exosome (CAF)-like properties in normal human breast fibroblasts by
cargos may also repress senescence as reported by van Balkom modulating the expression of genes involved in cell motility
et al. They found that endothelial cell-derived exosomes con- and extracellular matrix remodelling. In addition, they found
taining miR-214 may repress senescence by silencing the ex- that miR-9 inhibition could affect the migratory properties of
pression of a DNA damage-induced tumour suppressor, ataxia CAFs [122]. Others have found that exosomal transfer of hu-
telangiectasia mutated (ATM), in recipient endothelial cells to man telomerase reverse transcriptase (hTERT) mRNA from
stimulate migration and angiogenesis [121]. From the limited cancer cells to fibroblasts conferred several phenotypic chang-
studies available, it is becoming evident that targeting es to these fibroblasts, such as increased proliferation, delayed
exosomal transport to reverse dormancy and quiescence may senescence and resistance to DNA damaging drugs [123],
help to prevent the resurgence of tumours. Since the exosomal eventually leading to a sustained support of CAFs to tumour
contents may either activate or repress senescence, further growth and progression.
studies are warranted to delineate the exact role of exosome- Activation of signalling pathways is one of the mechanisms
induced senescence and to explore its potential for the devel- by which exosomes may modulate the tumour microenviron-
opment of novel therapeutic strategies. ment to promote tumour growth. Gu et al. reported exosomal
transfer of TGF-β and subsequent activation of the TGF-β/
4.2 Role of exosomes in modulating the tumour Smad pathway in human umbilical cord-derived mesenchy-
microenvironment and the formation mal cells (hucMSCs), through which gastric cancer cells can
of pre-metastatic niches trigger the differentiation of hucMSCs into CAFs. It was also
found that co-culture of hucMSCs with tumour cell-derived
4.2.1 Modulation of the tumour microenvironment exosomes increased the migratory capacity of hucMSCs and,
interestingly, that the treatment of cells with a TGF-β inhibitor
An important aspect of tumour progression is communication effectively blocked both hucMSC migration and their differ-
between tumour cells and cells within the tumour microenvi- entiation into CAFs [124]. These results reiterate the mecha-
ronment, such as fibroblasts, adipocytes and cells of the vas- nism underlying the modulatory effect of cancer cells on
cular and immune system (Fig. 2). Baroni et al. found, for tumour-associated cells favouring tumour growth. Moreover,
example, that miR-9 secreted via exosomes by breast cancer Webber et al. found that exosomal transfer of TGF-β from
cells have the ability to induce cancer-associated fibroblast cancer cells to fibroblasts resulted in their conversion to

Fig. 2 Schematic representation of exosome-mediated communication other proteins, may confer varying phenotypes, such as survival, evasion
between tumour cells and cells in the tumour microenvironment. from immune surveillance, metastasis and relapse, to tumour cells
Exosome-borne factors, such as microRNAs, receptors and a myriad of
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 233

myofibroblasts, as indicated by a strong induction of α-SMA expression of the translation initiation factors eIF4E and
[125]. Myofibroblasts present in tumour stroma play impor- eIF4GI may lead to a reduced migration and proliferation of
tant roles in the secretion of growth factors, such as activin A, cancer cells [139] and that exosomal miR-16-mediated down-
VEGF, insulin-like growth factor 1 (IGF-1) and, subsequently, regulation of VEGF may lead to inhibition of angiogenesis
inducing tumour cell proliferation, tumour progression, angio- [140] and downregulation of Akt [141]. MSCs may migrate
genesis and invasion [126–128]. Therefore, targeting the dif- to sites of injury [142] or tumour formation [143] in response
ferentiation of fibroblasts into myofibroblasts via exosomal to chemokines, such as stromal cell-derived factor 1 (SDF-1)
transfer may have important implications for the future de- [142], growth factors, such as basic fibroblast growth factor
signing of anti-cancer therapies. Bi-directional crosstalk be- (bFGF) [144], platelet-derived growth factor (PDGF), epider-
tween chronic myelogenous leukemia (CML) cells and stro- mal growth factor (EGF) [143] and VEGF [145]. Once MSCs
mal cells is known to play an important role in the survival and become part of the tumour microenvironment, they are ‘edu-
proliferation of CML cells by activation of EGFR signalling in cated’ through various factors including exosomes and, con-
stromal cells through amphiregulin that is present in CML- sequently, converted into tumour-associated MSCs. These
derived exosomes. EGFR signalling leads to IL8 upregulation tumour-associated MSCs can promote tumour growth
in stromal cells which favours CML cell survival. It has also through multiple mechanisms, including chemokine-
been found that pre-treatment of stromal cells with CML- mediated macrophage recruitment to tumour sites [146], ab-
derived exosomes may result in annexin A2 upregulation in errant miRNA expression resulting in downregulation of the
the stromal cells, which in turn facilitates the attachment of forkhead transcription factor FOXP2 to induce stem cell-like
leukemic cells to stromal monolayers [129]. Wu et al. reported features [147], interaction with endothelial cells to promote
that through activating the nuclear factor kappa B (NF-κB) angiogenesis [143], activation of various cellular signalling
pathway in macrophages, gastric cancer cell-derived pathways [148], modulation of immune responses [149] and
exosomes enhanced the secretion of pro-inflammatory cyto- increasing the population of CSCs [150], ultimately resulting
kines, which subsequently increased the proliferation and mi- in treatment failure due to the acquisition of drug resistance
gration of gastric cancer cells [130]. [151].
Also, tumour microenvironment-associated cells may se- Lin et al. showed that tumour-cell derived exosomes may
crete factors that facilitate tumour growth [131], and recent be internalised by bone marrow-derived MSCs and that this
work has shown that exosomes secreted by these cells may internalisation may subsequently alter these MSCs into tu-
not only play a major role in sustaining tumour growth, but mour promoting cells. These cells were found to favour tu-
also in acquiring drug resistance under hypoxic conditions. mour growth by promoting macrophage infiltration via en-
Languino et al. have, for example, reported transfer of TGF-β hanced production of CCR2 ligands in B16-F0 melanoma
receptor II (TβRII) from exosomes isolated from stromal fi- cells and EL-4 lymphoma cells in mice, and by increasing
broblasts of patients with OSCC to OSCC-associated the number of circulating monocytes. It was also found that
keratinocytes, which in turn led to increased TGF-β signal- treatment with a CCR2-specific inhibitor and macrophage ab-
ling in OSCC cells through re-activation of their response to lation by clodronate liposomes reversed the phenotype to nor-
TGF-β ligand [132]. Others have reported activation of mal MSCs [152]. The implication of MSC-promoted macro-
STAT1-dependent paracrine antiviral signalling in breast can- phage infiltration is an increase in monocyte-derived tumour-
cer cells through the transfer of exosomes from stromal cells, associated macrophages (TAMs). Monocytes generally sur-
and found juxtacrine NOTCH3 activation within the breast vive beyond their normal life span of two days under inflam-
cancer cells. They also found that the two pathways con- matory conditions in the tumour microenvironment and,
verged and that STAT1-mediated transcriptional changes re- hence, they may give rise to TAMs. Intriguingly, their survival
sulted in the establishment of drug-resistant breast cancer is mediated by cancer cell-derived exosome-induced inactiva-
cells [133]. Specialised adherent cells present in soft tissues tion of caspase-dependent apoptosis. MAPK pathway activa-
and body fluids such as bone marrow, adipose tissue, periph- tion through exosomal transfer of receptor tyrosine kinases,
eral blood, foetal liver, lung, amniotic fluid and umbilical such as phosphorylated EGFR and phosphorylated HER2,
cord blood are termed MSCs. These cells possess the ability may be favoured by this event [153].
to differentiate into mesenchymal tissue cells such as osteo- TAMs are important constituents of the tumour microenvi-
blasts, chondrocytes and adipocytes [134, 135]. MSCs can ronment and favour tumour progression by activating several
both suppress and enhance tumour growth. MSCs may sup- important events such as angiogenesis, drug resistance, im-
press tumour growth by inducing G1-phase cell cycle arrest mune evasion, activation of CSCs and promoting its self-re-
[136], dickkopf-1 (Dkk-1)-mediated inhibition of Wnt signal- newal, and extracellular matrix degradation [154]. These cells
ling [137] and reduction of IGF-1R/PI3K/Akt signalling by can exhibit either a M1 phenotype that suppresses tumour
sequestering free insulin-like growth factors (IGFs) [138]. In growth or a M2 phenotype that favours tumour growth [73].
addition, it has been found that downregulation of the Tumours with a high M2/M1 ratio usually exhibit a poor
234 V. Sundararajan et al.

prognosis, whereas polarization of TAMs towards a M1 phe- 4.2.2 From anti-oncogenic to pro-oncogenic
notype may increase the efficacy of anti-cancer drugs [154, exosome-mediated mechanisms
155]. A recent study has shown that TAMs in glioblastomas
may constitute 50% of the tumour mass, and that 85% of these Although different cells within the tumour microenvironment,
TAMs may be accounted for by infiltrating macrophages/ such as MSCs, TAMs and fibroblasts, may secrete exosomes
monocytes from the blood circulation. It was also found that that promote tumour growth, they may also possess intrinsic
bone marrow-derived monocytes may infiltrate the tumours anti-oncogenic functions. Only after they are recruited to tu-
during early stages in response to an increased secretion of the mour sites and reprogrammed by the cancer cells, they may
chemokine CCL2 by the tumour cells, and that these mono- favour tumour growth via their exosomal components. It has,
cytes may subsequently differentiate into macrophages and for example, been found that exosomes secreted by mouse
microglia-like cells. Inhibition of monocyte infiltration was embryonic fibroblasts (MEFs) may contain functional PTEN
found to improve survival in mouse models. Resident that, when internalised by recipient MEFs, may reduce the rate
microglial cells and TAMs exhibit differentially expressed of cell proliferation and the amount of pAKT [160]. In another
genes and it was found that among them cell migration was recent study, it was reported that treatment of A2780 and
most enriched in TAMs [156]. It has also been reported that SKOV-3 ovarian cancer cells with fresh or protease-digested
TAMs may favour ovarian tumour growth by inducing spher- exosomes derived from human adipose MSCs (hAMSC)-con-
oid formation via an increased adhesion between tumour cells ditioned media could downregulate the proliferation, as well
and TAMs resulting from EGF secretion by TAMs and acti- as the wound-repair and colony forming capacities, of these
vation of VEGF/vascular endothelial growth factor receptor cells, due to the presence of three potential new miRNAs,
(VEGFR) signalling through EGFR activation in the tumour seven potential candidate miRNAs and several known
cells. This activation was found to culminate in enhanced miRNAs in the exosomes, such as hsa-miR-4792, hsa-miR-
tumour cell proliferation, migration and trans-coelomic spread 320a, hsa-miR-320b, hsa-miR-7704, hsa-miR-181a-5p, hsa-
[157]. Recently, a role of exosomes in the polarization of miR-127-3p and hsa-miR-6087s. The ovarian cancer cells
macrophages towards the M2 phenotype was reported by were also found to be susceptible to apoptosis due to the fact
Shinohara et al., and they found that miR-145 present in that the exosomal miRNAs could upregulate pro-apoptotic
exosomes secreted by colorectal cancer cells may induce proteins, downregulate anti-apoptotic proteins and target im-
M2 macrophage polarization through histone deacetylase portant molecules associated with cell cycle progression and
11 (HDAC11) expression down-regulation and IL10 pro- survival pathways [161]. The anti-oncogenic role of exosomes
duction up-regulation. M2 macrophage polarization was secreted by hAMSC was further underscored by their ability
also found to cause significant increases in tumour vol- to upregulate the level of circulating and intra-tumoural natu-
umes in xenograft models co-injected with exosome- ral killer T-cells (NKT-cells) in HCC rat models, leading to an
treated macrophage-like cells [158]. increased anti-tumour activity and significantly smaller and
Multiple myeloma (MM)-derived exosomes may also es- lower grade-tumours compared to controls [162]. Another
tablish a favourable bone microenvironment for tumour anti-oncogenic function of mouse bone marrow-derived
growth by enhancing immunosuppression and regulating an- MSCs is the transfer of, next to other exosomal components,
giogenesis in bone marrow myeloid-derived suppressor cells miR-16, leading to inhibition of angiogenesis as a result of
(MDSCs) and bone marrow endothelial cells. These MM- decreased VEGF expression in recipient breast cancer cells
derived exosomes may modulate key angiogenic pathways [140]. Alcayaga-Miranda et al. reported that exosomes de-
via several angiogenesis-related proteins/regulators contained rived from menstrual stem cells (MenSCs) may exhibit anti-
in them , such as angiogenin, tissue inhibitor of oncogenic activity against PC3 prostate cancer cells resulting
metalloproteinase-1 (TIMP-1), Serpin E1, Serpin F1 and from a reduction in VEGF secretion, a decrease in reactive
VEGF-B. In addition, it was found that these exosomes could oxygen species (ROS) production, downregulation of
up-regulate the proliferation of MDSCs by activating the NF-κB expression and the suppression of pro-angiogenic fac-
STAT3 pathway (associated with self-renewal), and up- tors. Treatment of mice with MenSCs-derived exosomes led
regulating the pro-survival proteins Bcl-xL and Mcl-1. to in vivo regression of tumours exhibiting a decreased vascu-
These activated MDSCs exhibited enhanced inducible nitric lar density, reduced haemoglobin levels and reduced VEGF
oxide synthase (iNOS) production to favour the growth of and HIF-1α expression levels [163]. Human macrophages
MDSCs by suppressing the function of T-cells, thus facilitat- may also possess intrinsic anti-tumour activities before they
ing MM progression [159]. Taken together, these studies high- are recruited to tumour sites, as reported by Lee et al.
light a role of exosomes in mediating tumour-stroma commu- Specifically, they found that exosomal disintegrin and metal-
nication. Interfering with exosomal transfer may, hence, rep- loproteinase 15 (ADAM15) via its disintegrin-like domain
resent a novel strategy to target the tumour microenvironment may block interactions between integrin αvβ3 and
and, by doing so, to halt tumour progression. vitronectin, and inhibit vitronectin- and fibronectin-induced
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 235

growth and migration of breast, lung and ovarian cancer cells. responsible tor triggering these changes and that MIF inhibi-
The authors triggered the differentiation of monocytes to mac- tion abolished the preparation of pre-metastatic niches, as well
rophages and found that these monocyte-derived macro- as the metastasis of PDAC cells to the liver [165]. Hood et al.
phages abundantly secreted ADAM15 in their exosomes. reported that melanoma-derived exosomes homed to ipsilater-
These ADAM15+ exosomes were subsequently found to in- al sentinel lymph nodes in vivo and, subsequently, recruited
hibit the in vitro growth and migration of MDAH-2774 ovar- melanoma cells to these lymph nodes. Additional mechanistic
ian cancer cells, as well as to suppress their in vivo growth and studies revealed that melanoma-derived exosomes may acti-
to increase the survival of xenografted mice [164]. vate multiple metastatic pathways involved in basement mem-
brane invasion, migration, vascular organisation and the in-
4.2.3 Formation of pre-metastatic niches duction of angiogenic factors required for the survival and
growth of melanoma cells within lymph nodes [59, 169].
The establishment of pre-metastatic niches represents an Others reported on the role of differentially expressed
important phase in the metastatic process of cancer cells miRNAs in exosomes derived from bulk cancer cells and
in which exosomes play an important role [59]. Primary CSC sub-populations contributing to metastasis and pre-
tumour cells have been found to secrete several factors, metastatic niche formation in primary prostate cancers.
such as VEGF, TGF-β and lysyl oxidase (LOX), that Comparative exosomal miRNA profiling revealed 19
may recruit bone marrow-derived cells (BMDCs) to pre- miRNAs, including miR-100-5p (in both bulk and CSC
pare suitable microenvironments at secondary sites for the exosomes), miR-21-5p (in bulk exosomes) and miR-139-5p
attachment and survival of tumour cells [165–167]. The (in CSC exosomes) that may enhance prostate cancer prolif-
site of niche establishment depends upon various factors, eration and migration. Transfection of these miRNAs in nor-
including the expression and type of integrin present on the mal prostate WPMY-1 fibroblasts resulted in invasive and
exosome surface, the exosomal microRNAs content and metastatic characteristics through expression upregulation of
the presence of several supporting soluble factors. Below, MMP-2, -9, -13, as well as the receptor activator of nuclear
we will discuss the role of various exosomal factors in factor kappa-B ligand (RANKL) in the pre-metastatic niche,
preparing pre-metastatic niches, as well as in determining thereby promoting osteoclast recruitment and differentiation
the organ(s) of choice for metastasis. [170]. Peinado et al. found that melanoma cell-derived
Even though exosomes, independent of their origin exosomes could transform the phenotype of BMDCs to favour
from poorly or highly metastatic cells, are considered to metastasis by transfer of the MET oncoprotein. These
be the primary regulators and modulators that govern pre- exosomes could also induce vascular leakiness at pre-
metastatic niche formation, Jung et al. reported that this metastatic sites and educate BMDCs towards a pro-
event requires a soluble matrix, which depends on CD44v. vasculogenic phenotype [167].
Specifically, they found that CD44v6 expression knock-
down reduced the metastasizing capacity of pancreatic ad-
enocarcinoma cells and that this reduction could be res-
cued by supplementation of soluble matrix secreted by 5 Role of exosomes in cancer diagnosis
wild-type adenocarcinoma cells [168]. In another study, and treatment
it was found that exosomes released by human renal car-
cinoma cells expressing CD105 could promote angiogen- Although it is at present not fully understood how the sites
esis in normal endothelial cells. CD105+ exosomes could of pre-metastatic niches and, subsequently, metastasis are
also increase the adhesion of normal endothelial cells and, determined, Hoshino et al. provided new insights by focus-
by doing so, facilitate the formation of pre-metastatic ing on the expression of cell adhesion receptors, such as
niches in lungs by upregulating the expression of MMP- integrins, on exosomal surfaces. They noted that these
2, MMP-9, and VEGFR1 [67]. integrins may serve to prepare pre-metastatic niches by
Costa-Silva et al. reported, for the first time, a role of pan- interacting with components of the extracellular matrices
creatic ductal adenocarcinoma (PDAC)-derived exosomes in of the respective target organs. They found, for example,
the formation of pre-metastatic niches in the liver. An in- that exosomes expressing integrins α6β4 and α6β1 on their
creased production of TGF-β and an upregulation of fibronec- surface co-localised with S100A4+ cells in laminin-rich
tin production were seen after uptake of PDAC-derived lung environments to cause lung metastases, whereas
exosomes by Kupffer cells in the liver. The deposition of exosomes expressing integrins αvβ5 led to the formation
fibronectin stimulated the migration of BMDCs, such as mac- of liver metastases [171]. A further deciphering of the role
rophages and neutrophils, into the liver for the establishment of cell adhesion receptors on exosomes in organ-specific
of pre-metastatic niches. It was found that macrophage migra- metastasis may hold promise for the development of new
tion inhibitory factor (MIF) was the exosomal component therapeutic strategies.
236 V. Sundararajan et al.

5.1 Exosome signatures for diagnosis and treatment Molecular exosome profiling can also be used to predict
responses to anti-cancer therapies, such as docetaxel in the
Since exosomes may carry a variety of biomolecules, such as treatment of castration-resistant prostate cancer (CRPC). It
mRNAs, miRNA and proteins, and since these biomolecules has been found that DU145 prostate cancer cells that are either
can be isolated from various body fluids with ease, it has been sensitive (DU145 Tax-Sen) or resistant (DU145 Tax-Res) to
suggested that they may be used as diagnostic/prognostic sig- docetaxel differ in the amounts of exosomes secreted.
natures using minimally invasive techniques [172]. Chen et al. Exosomes isolated from DU145 Tax-Res cells were addition-
found, for example, that of 107 proteins that were differential- ally found to be enriched in drug efflux proteins such as
ly expressed, 24 differed significantly in urinary exosomes MDR-1 and MDR-3. The same was observed in sera of
isolated from bladder cancer patients and non-cancer patients. CRPC patients and it was found that the presence of these
They also found that urinary exosome-derived calcium-signal proteins was indicative of resistance to docetaxel treatment
transducer 2 (TACSTD2), a cell surface glycoprotein that is [180]. Similarly, integrin β4 and vinculin levels were found
overexpressed in numerous cancers, may serve as a biomarker to be elevated in exosomes isolated from taxane-resistant PC-
for the diagnosis of bladder cancer. Specifically, they found 3 prostate cancer cells and could be used as exosome-based
that TACSTD2 levels were higher in microparticles secreted diagnostic tools to predict taxane resistance in prostate cancer
by bladder cancer patients compared to those secreted by con- patients [181].
trols, i.e., hernia and urinary tract infection cases [173]. Others The amount of exosomes released in blood and their pro-
have reported that loss of two proteins that regulate mitochon- tein content may differ between healthy and cancer patients,
drial function (SH3GL2 and MFN2) in serum exosomes cor- which can be used as a diagnostic tool. It has, for example,
related with the presence of breast cancer and the occurrence been found that plasma from patients with chronic lymphoid
of lymph node metastases [174]. The diagnosis of prostate leukaemia (CLL) may have significantly increased exosome
cancer is challenged by unnecessary biopsies in patients with levels, and miRNA profiling revealed a CLL-specific signa-
benign disease and unequivocal prostate specific antigen ture characterised by miR-150, miR-155 and miR-29a-c up-
(PSA) levels. To overcome these challenges, exosomal regulation [182]. It has also been revealed by plasma-derived
RNAs were isolated from urine after which a novel molecular exosome analysis of melanoma patients of different stages that
signature, called EXO106 score, was deduced using the sum stage IV disease patients may exhibit a higher content of sev-
of normalised PCA3 and ERG RNA levels. Using the eral (onco)proteins that correlate with a poor survival [167].
EXO106 score, fewer biopsies were needed for men with Others found that high exosome levels in plasma from colo-
unequivocal PSA scores, i.e., the EXO106 score provided a rectal cancer patients was associated with poorly differentiated
negative predictive value of 97.5% for the diagnosis of high- tumours and high carcinoembryonic antigen (CEA) plasma
grade prostate cancer [175]. He et al. performed proteomic levels [183]. Taken together, it can be concluded that exosome
analyses of three HCC-derived cell lines and found that levels and their contents may be associated with multiple dis-
metastasis-inducing factors, such as MET, S100 family mem- ease parameters. As such, they may be used as non-invasive
bers and caveolins, were present only in exosomes derived diagnostic/prognostic signatures.
from metastatic HCC cells [176].
Cancer may also be diagnosed through the measurement of
miRNA and mRNA levels in blood-derived exosomes [73]. 5.2 Exosomes: potential diagnostic biomarkers
Joshi et al. found, for example, that high levels of miRNA-10b
isolated from plasma-derived exosomes corresponded to the As mentioned above, exosomes can be isolated from a variety
occurrence of pancreatic cancer and/or chronic pancreatitis of body fluids such as serum, plasma, saliva, urine and cere-
[177]. Others revealed, through miRNA profiling of serum- brospinal fluid (CSF). Biomolecules contained within these
derived exosomes from glioblastoma multiforme (GBM) pa- exosomes include cell surface glycoproteins, double stranded
tients, a diagnostic signature consisting of either the small DNA, miRNAs, lncRNAs, drug efflux proteins and other
noncoding RNA RNU6-1 alone or a combination of miR- proteins that are associated with various processes of cancer
320, miR-574-3p and RNU6-1 that may differentiate GBM development and progression. The levels of these biomole-
patients from healthy individuals [178]. Similarly, exosomal cules may be either upregulated or downregulated, depending
signatures have been identified for the diagnosis and monitor- on the stage and aggressiveness of the cancer, compared to
ing of melanoma [167] and breast cancer [84] patients. Skog those of healthy subjects. This information may facilitate the
et al. reported that mRNA mutants and glioblastoma-specific non-invasive diagnosis of cancer [73]. In addition, the respec-
miRNAs can be detected in serum-derived exosomes of GBM tive biomolecules may be employed to monitor disease recur-
patients. Specifically, they found that EGFRvIII variant rence and response to therapy [73]. Several of these biomol-
mRNA could be detected and used as a non-invasive diagnos- ecules and their diagnostic/prognostic significances are listed
tic marker [179]. in Table 1.
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 237

Table 1 Exosomal biomolecules and their clinical significance

Type of cancer Biomolecule Exosome source Clinical significance Reference

Acute myeloid leukaemia CD34 Plasma Presence of elevated levels of CD34(+) [213]
exosomes in cancer patients
Bladder cancer EDIL-3 Urine Increased EDIL-3 levels in high grade [214]
bladder cancer patients alone. EDIL-3
favours angiogenesis and endothelial
cell migration by EGFR signalling
lncRNAs HOTAIR, HYMA1, Urine Urinary exosomes of patients with [215]
LINC00477, LOC100506688, high-grade muscle-invasive disease
and OTX2-AS1 enriched in these lncRNAs
Breast cancer miR-101 and miR-373 Serum Elevated levels of miR-373 in triple [216]
negative, ER(−) and PR(−) cancer
patients. miR-101 increased only in
breast cancer patients when compared to
benign and healthy cases
Survivin Serum Survivin and its splice variant [217]
survivin-ΔEx3 were elevated in all
cancer patients, whereas survivin-2B
exhibited a differential expression in
breast cancer patients
Periostin Plasma Compared to patients with localised [218]
disease, patients with lymph node
metastasis have higher levels of
exosomal periostin
Cervical cancer miRNA-21 and miRNA-146a Cervicovaginal lavage Abnormally elevated only in cervical [219]
cancer patients
Colon cancer miR-19a Serum Elevated expression in cancer patients [220]
alone. High expression was correlated
with a poor prognosis and tumour
recurrence
miRNAs let-7a, miR-1229, Serum Compared to healthy controls, colon [221]
miR-1246, miR-150, miR-21, cancer patients had increased levels of
miR-223, and miR-23a these miRNAs
Gastric cancer LINC00152 Plasma Elevated levels in gastric cancer [222]
patients alone
miR-21 and miR-1225-5p Peritoneum lavage fluid Highly elevated in T4 metastatic stage [28]
patients compared to T1-T3 stages.
High expression was correlated with
peritoneal recurrence after gastric
cancer resection
Glioma miR-21 Cerebrospinal fluid CSF of glioma patients contained high [26]
levels of miR-21. Its expression was
correlated with metastasis
and recurrence
Hepatocellular carcinoma miR-21 Serum miR-21 levels were elevated in patients [223]
with HCC compared to healthy
volunteers and chronic hepatitis B
patients. Higher levels were correlated
with cirrhosis and advanced tumour
stage
miR-718 and miR-1246 Serum Downregulation of these miRNAs were [224]
correlated with recurrence after liver
transplantation. Decreased miR-718
levels were associated with
tumour aggressiveness
Laryngeal squamous miR-21 and HOTAIR Serum Higher expression in patients with LSCC [225]
cell carcinoma and lymph node metastasis than in those
with benign disease
Melanoma MDA-9 and GRP78 Serum Only metastatic melanoma patients [226]
exhibited elevated expression levels of
these markers
miR-125b Serum Significantly downregulated in patients [227]
with advanced melanoma compared to
238 V. Sundararajan et al.

Table 1 (continued)

Type of cancer Biomolecule Exosome source Clinical significance Reference

disease-free melanoma patients and


healthy controls
Nasopharyngeal Galectin-9 Serum Exosome-derived galectin-9 was detect- [228]
carcinoma able only in cancer patients
Non-small cell LRG1 Urine LRG1 expression was significantly [229]
lung cancer up-regulated in NSCLC patients com-
pared to controls
Oesophageal squamous miR-21 Serum Elevated only in ESCC patients; correlated [230]
cell carcinoma positively with tumour progression
and aggressiveness
Ovarian cancer miR-21, miR-141, miR-200a, Serum Elevated levels of these 8 miRNAs [231]
miR-200c, miR-200b, miR-203, in sera of ovarian cancer patients
miR-205, and miR-214 compared to controls
miR-222-3p Serum Higher levels in sera of cancer patients [232]
compared to controls
Pancreatic cancer Double-stranded genomic DNA Serum Compared to healthy controls, only [233]
with mutated KRAS and p53 pancreatic cancer patients exhibited
mutations in KRAS and p53 DNA
Glypican-1 (GPC1) Serum GPC1(+) exosome levels were higher in [234]
cancer patients than in healthy control
and benign prostatic disease patients.
Patients with distant metastases
exhibited higher GPC1(+) exosome
levels compared to those with lymph
node metastasis or no metastasis
Prostate cancer PSA, PSMA Urine Present only in cancer patients [235]
P-glycoprotein Serum Elevated levels in docetaxel-resistant [236]
prostate cancer patients
miR-1290 and miR-375 Plasma Higher levels were correlated with a poor [237]
overall survival in castration-resistant
prostate cancer patients

6 Exosome-based targeted therapy drug cyclophosphamide in mouse models. In addition, they


found that treatment with DMA or omeprazole inhibited the
6.1 Inhibition of exosome secretion and transfer phosphorylation of STAT3 in MDSCs, thereby reducing its im-
of oncogenic molecules munosuppressive ability and its ability to elicit immune evasion
of cancer cells [186].
As discussed above, SMase has been found to be involved in the Also, Rab GTPases that play important roles in exosome
secretion of exosomes by regulating the synthesis of ceramide. secretion (see above) and other proteins that are known to be
Kosaka et al. found that GW4689, a chemical inhibitor that involved in exosome secretory pathways have been tested as
targets SMase, could reduce the secretion of miRNAs [184]. It putative therapeutic targets. Using a mouse model, Bobrie
was also found that GW4689-based exosome secretion targeting et al. found that Rab27 inhibition resulted in reduced exosome
effectively reduced the in vivo occurrence of lung metastases in secretion. As a consequence, they observed a decrease in pri-
mice injected with Lewis lung carcinoma (LLC) cells. Exosomes mary 4T1 breast cancer growth and its metastasis to lungs. In
derived from injected LLC cells could overcome this inhibition, addition, they found that Rab27a-dependent secretion of
thereby underscoring the importance of exosome-mediated can- exosomes resulting in the mobilization of neutrophils was
cer promotion [185]. Since intracellular calcium levels may play required for 4T1 tumour growth [187]. A recent study showed
important roles in exosome secretion (see above), the targeting of that inhibition of YKT6, a SNARE protein that is involved in
calcium channels to modulate exosome secretion has been in- exosome synthesis and secretion, using siRNA and pre-
vestigated as a therapeutic option. The effect of inhibiting H+/ miRNAs, led to a reduction in exosome secretion by approx-
Na+ and Na+/Ca2+ channels, using dimethyl amiloride (DMA), a imately 80%. Since YKT6 expression is inversely correlated
drug that blocks exosome formation, and K+/H+ ATPase using with the survival of non-small cell lung cancer (NSCLC) pa-
omeprazole, on exosome secretion has been studied by Chalmin tients, inhibiting its activity and decreasing exosome synthesis
et al. They found that both the drugs reduced exosome secretion may have important clinical implications [188]. Through a
in vitro and in vivo, and enhanced the efficacy of the anti-cancer comparative proteome analysis of exosomes secreted by
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 239

normal human T-cell blasts and acute leukaemia Jurkat T- inhibitors to suppress cell-cell communication and subse-
cells, it was found that Jurkat cell-derived exosomes were quent anti-cancer drug administration to potentiate the che-
enriched in 14 membrane proteins, the most abundant being motherapeutic effect. The latter approach seems to be more
valosin-containing protein (VCP). VCP is a membrane effective than the former, since it inhibits the communica-
ATPase that acts to maintain endoplasmic reticulum (ER) ho- tion between cancer cells as well as between cancer cells
meostasis and ubiquitination, and may serve as a prognostic and other cells in the tumour microenvironment which, in
biomarker for gastric carcinoma. Treatment with a VCP inhib- turn, may sensitise cancer cells to chemotherapy.
itor (DBeQ) was found to result in inhibition of exosome
secretion only in Jurkat cells. Based on their results, the au- 6.2 Exosome-based immunotherapy
thors concluded that VCP targeting may be a novel therapeutic
approach that interferes with exosome release [189]. In recent years, exosomes have gained interest for their ability
It has also been reported that inhibition of exosome secre- to modulate immune functions, especially exosomes derived
tion may help to overcome drug resistance. Next to a low pH, from dendritic cells (DCs). These DC-derived exosomes (DC-
cisplatin resistance in human melanoma cells was, for exam- Exo) carry antigens that can activate the immune system and
ple, found to be due to the secretion of cisplatin-containing stimulate the activity of immune cells, such as CD8+ T-cells,
exosomes. Treatment of these cells with the proton pump in- which may lead to tumour eradication. In addition, it has been
hibitor (PPI) lansoprazole resulted in an enhanced uptake of found that tumour cells, when co-cultured with DC-Exo, may
cisplatin and an inhibition of exosome release. In addition, it exhibit enhanced abilities to activate T-cells [194, 195].
was found that tumours obtained from xenograft models DCs pulsed with tumour-derived exosomes (TEXs) may
contained more cisplatin when the animals were exposed to exhibit increased efficacies in eliciting immune responses
PPI and cisplatin. Subsequent plasma analysis revealed a de- both in in vitro and in vivo models. Rao et al. showed, for
crease in the number of exosomes as well as in their cisplatin example, that DCs pulsed with hepatocellular carcinoma
content [190]. Recently, Li et al. found that exosomes may (HCC) TEXs exhibited potent immune responses in in vitro
contribute to drug resistance when tyrosine kinase inhibitors and in vivo mouse models by modulating the tumour immune
(TKIs), such as gefitinib, and chemotherapeutic agents, such microenvironment. In orthotopic models, significant tumour
as cisplatin, were co-administered. Gefitinib-treated PC9 suppression was observed and improvements in immune func-
NSCLC cells may acquire cisplatin resistance by upregulating tions were evident based on increased numbers of T lympho-
autophagy and downregulating key apoptotic proteins. The cytes, increased interferon-γ and decreased IL10 and TGF-β
authors also found that GW4869-mediated inhibition of expression levels at tumour sites [196]. Anti-tumour immune
exosome secretion in gefitinib-treated NSCLC cells not only functions elicited by exosomes can be further enhanced
reduced antagonistic effects when TKIs were co-administered by the addition of ligands to tumour cells that stimulate
with cisplatin, but also induced synergistic effects [191]. DCs by using exosomes secreted by tumour cells. Wang
Other studies have been aimed at suppressing the pro- et al. used exosomes derived from CD40 ligand gene-
tumorigenic effects of exosomes by natural compounds and modified lung tumour cells (CD40L-Exo) and found that
nutraceuticals. Zhang et al., for example, found that curcumin these exosomes promoted DC maturation and upregulated
could reverse exosome-mediated inhibition of NK cell cyto- the secretion of cytokines, such as interferon-γ and IL12,
toxicity. Specifically, they found that exosomes isolated from in mouse splenocytes. In addition, they observed an in-
TS/A breast cancer cells could inhibit IL2-induced NK cell creased ex vivo proliferation of CD4+ T-cells and that DCs
cytotoxicity by inhibiting the activation of STAT5. Treatment pulsed with CD40L-Exo effectively inhibited tumour
with curcumin caused a dose-dependent increase in growth and increased the survival of mice inoculated with
ubiquitinated exosomal proteins and Jak3-mediated phos- LLC cells [197]. Another way to improve the efficacy of
phorylation of STAT5 [192]. Interestingly, curcumin and its DC-Exo is by culturing them in the presence of DC mat-
derivatives are known to target not only the cancer cells, but urating agents. Such matured DCs are more effective in
also CSCs through multiple regulatory mechanisms, hence eliciting anti-tumour responses than immature DCs. Damo
sensitising resistant populations for a better treatment efficacy et al. reported the use of DC-Exo loaded with antigens
[193]. Taken together, curcumin may be able to reverse im- and poly I:C as a DC maturating agent along with oval-
munosuppressive effects induced by tumour cell-derived bumin (OVA) to boost anti-tumour immunity in mouse
exosomes and increase the sensitivity of cancer cells to stan- melanoma models. The DC-Exo vaccines thus obtained
dard chemotherapy. were found to be more effective in stimulating OVA-
So, cancer cell sensitisation towards chemotherapy may specific CD8+ and CD4+ T-cells to acquire effector func-
be achieved via two approaches: (i) concomitant adminis- tions [198]. Heat shock proteins (HSPs), which represent
tration of drugs that inhibit exosome secretion and anti- important constituents of exosomes and serve as stress-
cancer drugs and (ii) initial administration of exosome induced molecular chaperones [199], can stimulate
240 V. Sundararajan et al.

immune functions by augmenting the ability to target can- effective and biocompatible means of producing exosomes
cer cells. Li-Hong et al. reported that exosomes contain- using bovine milk. Different drugs, such as withaferin A
ing hsp60, hsp70 and hsp90 secreted by drug-resistant (WFA), bilberry-derived anthocyanidins, curcumin, paclitaxel
HepG2 HCC cells increased the activity of cytotoxic NK and docetaxel were encapsulated in the exosomes. By doing
cells, upregulated the expression of the inhibitory receptor so, they found that drugs loaded in bovine milk-derived
CD94 and downregulated the expression of the activating exosomes exhibited increased efficacies compared to the free
receptors CD69, NKG2D and Nkp44 [200]. In addition, it drugs and Exo-WFA was found to be effective in in vitro lung
has been found that the location of HSPs in exosomes and breast cancer cell models, as well as in in vivo xenograft
plays a crucial role in determining their anti-tumour im- lung cancer models. It turned out that the exosomes remained
munity. It has, for example, been found that exosomes in circulation until day 6. In addition, it was found that animals
derived from myeloma cells with membrane bound treated with empty exosomes did not exhibit any abnormal
Hsp70 may exhibit enhanced DC maturation capacities effects, indicating the safety of bovine milk-derived
[199]. Thus, exosomes derived from DCs and other exosomes. Conjugation of folate to the exosomes further in-
sources may serve as excellent vaccines for immunother- creased the tumour targeting ability of Exo-WFA [204]. In
apy, and clinical trials have already been initiated to ex- order to overcome limitations associated with exosome pro-
plore their safety and efficacy in humans (see below). duction and scalability, Jang et al. synthesised exosome-
mimetic nanovesicles by the breakdown of monocytes/
6.3 Engineered exosomes as therapeutic cargos macrophages for targeted delivery of anti-cancer drugs.
These exosome-mimetics had a higher production yield
In recent years, exosomes have also received considerable (100-fold). Through in vitro studies it was found that these
attention as putative drug delivery vehicles due to several nanovesicles loaded with doxorubicin retained their ability
unique properties that they possess. Since exosomes are natu- to selectively deliver drugs and induce tumour necrosis factor
rally derived vesicles, they do not trigger undesirable immune alpha (TNF-α)-stimulated endothelial cell death. In a mouse
reactions and they remain undetected by the complement sys- tumour model, the exosome mimetics were found to be effec-
tem, thus providing them with a better stability [201]. tive in reducing tumour growth without causing any adverse
Recently, Aqil et al. investigated the efficacy of celastrol side effects [205].
(CEL), a natural compound encapsulated in exosomes, against As of yet, exosomes have been successfully used as cargos
NSCLC cells. They found that CEL in a time- and for the delivery of miRNAs, anti-miRNAs and siRNAs to
concentration-dependent manner inhibited the proliferation inhibit tumour growth and drug resistance. Breast cancer
of both lung carcinoma A549 (IC50 1.8 μM) and H1299 Hs578T cells and its aggressive triple negative clonal variant
(IC50 1.4 μM) cells. Mechanistically, they found that CEL Hs578T(i)8 exhibit enhanced invasion, migration and resis-
inhibited TNFα-induced NF-κB activation, upregulated the tance to cisplatin due to miR-134 downregulation. It was
expression of ER stress chaperones and induced apoptosis. found that exosome encapsulated miR-134 delivered to these
They also noted that exosome encapsulated CEL exhibited breast cancer cells reduced their migrative and invasive capac-
significant anti-tumour activity in xenograft models without ities, and sensitised them to anti-hsp90 drugs. Upon miR-134
systemic toxicity [202]. Others investigated an exosomal for- expression in the cells, significant decreases in STAT5B,
mulation of paclitaxel (ExoPTX) to overcome multidrug re- Hsp90 and Bcl-2 protein levels were found [206]. Ohno
sistance in cancer cells. Exosomes shed by macrophages were et al. investigated the effect of let-7a miRNA-containing
used and subsequent encapsulation of paclitaxel produced exosomes decorated with GE11 peptide to target EGFR ex-
ExoPTX with a high loading capacity and a sustained release pressing breast cancer cells through an EGFR-dependent in-
rate. By using a drug-resistant kidney cell line (MDCKMDR1) ternalization process. They injected luciferase-expressing
it was found that ExoPTX was 50 times more cytotoxic. HCC70 tumour cells into mammary fat pads of RAG2−/− mice
Importantly, the authors found that encapsulation of paclitaxel and found that, compared to control exosomes, the GE11 dec-
in exosomes enabled it to bypass the P-gp efflux protein either orated exosomes bound 3 times more efficiently to the tumour
by endocytosis-mediated transport or by fusion with the plas- cells. GE11 decorated exosomes containing let-7a miRNA
ma membrane. In LLC-derived pulmonary metastatic mouse also significantly inhibited HCC70 tumour growth, but the
models, intranasal administration of ExoPTX resulted in its genes that are normally repressed upon let-7a expression were
localization to cancer cells and a significant inhibition of the found to be unaffected. The authors concluded that let-7a may
growth of lung metastases [203]. act by modulating other, as yet unknown, genes [207]. In
Some of the work related to exosomes as drug delivery another study, exosomes obtained from human embryonic
vehicles has been aimed at strategies to produce exosomes at kidney 293 (HEK293) cells were used to deliver siRNA
large scale that are devoid of harmful side-effects. In a recent against polo-like kinase 1 (PLK-1) in UMUC3 bladder cancer
study Mungala et al. investigated options to develop cost cells. PLK-1 is an important regulator of mitotic cell cycle
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 241

progression and its upregulation has been found to result in Taken together, these studies highlight the efficacy of
increased tumour recurrence and metastasis. Delivery of this exosomes as novel drug delivery vehicles. As such, they may
siRNA resulted in PLK-1 mRNA and protein expression be instrumental for the design of targeted therapeutic approaches.
knockdown and, concomitantly, apoptosis and necrosis induc-
tion [208]. In temozolomide (TMZ)-resistant glioblastoma
cells it was found that anti-miR9 delivery via MSCs by either
7 Clinical Trials
gap junction communication or by exosome transfer down-
regulated the expression of MDR-1, thereby causing a reversal
Extensive research regarding the role of exosomes in cancer
of drug resistance and a re-sensitisation to TMZ [209].
development and progression, as well as their putative rele-
In order to improve the targeting abilities of exosomes and to
vance as diagnostic and/or therapeutic targets, has led to their
overcome limitations such as large scale production and iden-
inclusion into a variety of clinical trials. Most of the trials that
tification of suitable cell types for the production of exosomes,
are currently active or completed have focussed on the use of
Qi et al. developed a dual-functional exosome-based
exosomes as drug delivery vehicles for cancer therapy, the
superparamagnetic nanoparticle cluster for drug delivery. In
identification of biomarkers for cancer diagnosis and
the presence of an external magnetic field, these exosomes
exosomes derived from dendritic cells as cancer vaccines.
(SMNC-EXO) exhibit superparamagnetic properties at room
No adverse effects or severe toxicities have been reported in
temperature enabling their efficient targeting to cancer cells.
the clinical trials conducted so far. Based on current knowl-
SMNC-Exo was found to exhibit excellent stability in fresh
edge, exosomes hold promise as cancer biomarkers, drug car-
serum and to be biocompatible without adverse side effects in
go vehicles and cancer vaccines. In Table 2 several clinical
mice. SMNC-EXO loaded with doxorubicin (D-SMNC-EXO)
trials that are currently active, completed and/or not yet open
accumulated passively in vivo at tumour sites to release doxo-
for recruitment are listed.
rubicin via the enhanced permeability and retention (EPR) ef-
fect. The application of a magnetic field favoured active
exosome accumulation. Among different treatment groups test-
ed, mice dosed with D-SMNC-EXO in the presence of a mag- 8 Conclusions and future perspectives
netic field exhibited the highest tumour suppressive activities
by decreasing Bcl-2 expression and increasing caspase-3 ex- In spite of being a relatively new field of research, exosomes
pression [210]. To further reduce toxicity, Tian et al. used im- have gained ample interest due to their multifaceted role in
mature mouse dendritic cells (imDCs) for exosome production. tumour biology, and their perspective as comprehensive
These exosomes were engineered to express Lamp2b fused to tools for cancer eradication. Since exosomes mimic the con-
an iRGD peptide specific for αγ integrin and loaded with doxo- tents of the tumour cells from which they originate, they
rubicin (iRGD-Exos-Dox). The authors found that these possess the ability to affect neighbouring tumour cells as
exosomes had a high encapsulation efficiency and delivered well as distant tumour and normal cells via circulation in
the drug to αγ integrin-positive breast cancer cells with an different body fluids. It has been well documented now that
efficiency of approximately 95%. It was also found that transfer of exosomal components, such as mRNAs,
iRGD-Exos-Dox inhibited the proliferation of different cancer miRNAs, lipids, proteins, signalling/epigenetic regulators
cell types and delivered the drug efficiently to tumours in a and DNA molecules, may affect the behaviour of recipient
triple-negative breast cancer mouse model. Mice treated with cells. This exosome-mediated transfer of molecules may, for
control exosomes exhibited a 15-fold increase in tumour vol- example, result in uncontrolled proliferation, epithelial-
ume, whereas iRGD-Exos-Dox treated mice exhibited only a 4- mesenchymal transition, angiogenesis and distant metastasis
fold increase in tumour volume, with no mortality, morbidity, through modulation of the tumour microenvironment, expul-
cardiotoxicity, hepatotoxicity and/or damage to other organs, sion of anti-cancer drugs to cause drug resistance, prepara-
such as spleen and lungs, thus suggesting a potential improve- tion of pre-metastatic niches, enhanced migration of tumour
ment in therapeutic efficacy [211]. cells and re-emergence of tumours at metastatic sites after a
Therapeutic approaches aimed at delivering drugs to ma- prolonged period of dormancy. Together, these phenomena
lignant conditions in the brain often fail because of their in- impose major challenges on cancer treatment. Apart from its
ability to cross the blood-brain barrier (BBB). To overcome tumour promoting effects, exosomes have also been found
this problem, exosomes isolated from brain endothelial cells to serve as potential diagnostic/prognostic biomarkers.
were used as vehicles to deliver paclitaxel or doxorubicin to Current knowledge enables their use, not only as biomarkers
brain cancer cells. It was found that the drugs encapsulated in for early cancer diagnosis, but also as useful biomarkers for
endothelial cell-derived exosomes efficiently penetrated the predicting anti-cancer drug responses. The high stability of
BBB and resulted in smaller U-87 MG tumour masses in biomarkers within exosomes turns them into particularly
xenografted zebrafish embryos [212]. suitable tools for these purposes. It is anticipated that these
242 V. Sundararajan et al.

Table 2 Summary of completed, active and yet to be completed clinical trials using exosomes for the diagnosis and treatment of cancer

Clinical trial Status Result Reference

Drug delivery & therapeutics: Unknown – [238]


Phase I clinical trial of the ability of plant
exosomes to deliver curcumin to normal and
colon cancer tissue
Investigation of the ability of grape exosomes Recruiting – [239]
to prevent oral mucositis in patients with head
and neck cancer undergoing chemo- and ra-
diotherapy
Immunotherapy: Completed 32% of the patients exhibited disease stabilization of [240]
Phase II clinical trial to investigate the effect of more than four months; an increase in NK cell
second generation DC-derived exosomes function was found in some patients with
(Dex) (IFN-γ-Dex) loaded with MHC class I defective NKp30 expression
and class II restricted cancer antigens to boost
NK and T cell immune responses in inoperable
NSCLC patients after first line chemotherapy
Phase I clinical trial to investigate the Completed Dex enhanced the number of circulating NK cells [241]
mechanism of activation of NK cells by Dex in and restored NKG2D ligand expression in
melanoma patients circulating T and NK cells to elicit
NKG2D-dependent NK cell cytotoxicity in 7/14
patients. NKG2D ligand present on the surface
was required for NK cell activation. IL-15Rα was
important for NK cell proliferation and IFN-γ
secretion by NK cells
Phase I clinical trial to investigate the safety Completed No grade II toxicity was found and the maximum [242]
and feasibility of autologous exosomes pulsed tolerated dose was not achieved. According to the
with MAGE3 peptide as immunotherapy in RECIST criteria, one patient exhibited a partial
stage III/IV response. Large scale exosome production was
melanoma patients feasible and exosome administration was safe
Diagnosis: Completed Not available [243]
To correlate urinary exosome gene signatures
with the presence or absence of high grade
prostate cancer in prostate needle biopsies
Investigation of gastric cancer-derived Recruiting – [244]
exosomes as biomarkers and to study the
prognostic efficacy of such exosomes in gas-
tric cancer patients
Pilot study investigating the use of exosomes Recruiting – [245]
as a screening modality to diagnose
oropharyngeal squamous cell carcinoma in
human papilloma virus-affected patients
Pilot study to investigate whether Hsp70 Recruiting – [246]
exosomes in blood and urine samples of
patients with malignant solid tumours may be
used as diagnostic biomarkers
Analysis of urinary exosomal proteins to Not yet open for recruitment – [247]
identify prognostic markers in patients with
papillary, follicular or anaplastic thyroid
cancer before surgery, immediately after
surgery and at different time points
post-surgery
Safety and efficacy: Completed Not available. However, based on the favourable [248] [249]
Phase I clinical trial to investigate the safety of safety profile, currently participants are recruited
autologous malignant glioma cells derived for a dose escalation study
from malignant glioma patients treated with
insulin-like growth factor receptor-1 antisense
molecules encapsulated in diffusion chambers
implanted in rectus sheath and exosomes re-
leased from dying tumour cells in activating
the immune system against the tumour
Phase I clinical trial to investigate the efficacy Completed Combination of Aex with GM-CSF was safe, well [250]
of ascites-derived exosomes (Aex) combined tolerated and induced anti-tumour cytotoxic T
with granulocyte-macrophage lymphocyte responses
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 243

Table 2 (continued)

Clinical trial Status Result Reference

colony-stimulating factor (GM-CSF) as im-


munotherapy in colorectal cancer patients
Pilot study investigating the effect of the Recruiting – [251]
BRAF inhibitor vemurafenib on exosomes
produced by patients with advanced
unresectable or metastatic melanoma
Molecular studies: Recruiting – [252]
Evaluation of miRNA expression in serum,
bile, oesophageal cells, and miRNA
expression within biliary exosomes to
differentiate between gastroesophageal reflux,
Barrett’s oesophagus and cancer
To investigate the consistency of PD-L1 ex- Not yet open for recruitment – [253]
pression in cancer tissue and plasma exosome
in NSCLC patients
To investigate the consistency of PD-L1 ex- Not yet open for recruitment – [254]
pression in lung cancer tissues and plasma
exosomes before and after radiotherapy at
different time points to probe the best timing at
which PD-L1 is expressed
Disease recurrence and response to treatment: Recruiting – [255]
Role of exosome-mediated intercellular sig-
nalling and disease recurrence in pancreatic
cancer patients
To investigate the efficacy of high dose Recruiting – [256]
hypofractionated radiotherapy in different
cancers by quantifying the number of immune
cells, as well as the amount of secreted factors
and exosomes released in blood before, during
and after high
dose radiotherapy

approaches will improve future cancer care and foster better effective protocols, prolonged circulating times and biocom-
treatment outcomes. patible exosomes, further controlled pre-clinical and clinical
The ability of exosomes to carry a diverse range of mole- studies are warranted.
cules has enabled their use as depots to deliver conventional
anti-cancer drugs, as well as natural compounds, miRNAs, Acknowledgements This work was supported by University of Malaya
Programme Grant RP032-14HTM. The authors would like to thank Miss
siRNAs and anti-miRNAs. In vitro and in vivo studies have
Yew Hong Wen, from Stem Cell Biology Laboratory, for her assistance in
provided promising results and they have laid a foundation for preparing the artwork and figures.
ongoing and future clinical trials. Another promising aspect is
the use of exosomes as vaccines for the treatment of cancer. Compliance with ethical standards
Exosomes derived from dendritic cells and other sources have
shown the ability to boost the capacity of immune cells to Conflict of interest The authors declare that they have no conflict of
eradicate tumour cells, and they are currently in clinical trials interest.
to test their safety.
Though several studies performed so far have succeeded in
using exosomes for cancer diagnosis, prognosis and therapy, it
is important for future studies to take into account some of the References
challenges that still lie ahead related to large scale exosome
production, biocompatibility, biodistribution and the efficient 1. S.E.L. Andaloussi, I. Mager, X.O. Breakefield, M.J. Wood,
Extracellular vesicles: biology and emerging therapeutic opportu-
isolation of exosomal biomarkers from body fluids. It is also nities. Nat. Rev. Drug Discov. 12, 347–357 (2013)
important to mention here that although some studies have 2. R. Kalluri, M. Zeisberg, Fibroblasts in cancer. Nat. Rev. Cancer 6,
already reported large scale exosome production, cost 392–401 (2006)
244 V. Sundararajan et al.

3. D.D. Yu, Y. Wu, H.Y. Shen, M.M. Lv, W.X. Chen, X.H. 20. B.T. Pan, R.M. Johnstone, Fate of the transferrin receptor during
Zhang, S.L. Zhong, J.H. Tang, J.H. Zhao, Exosomes in devel- maturation of sheep reticulocytes in vitro: selective externalization
opment, metastasis and drug resistance of breast cancer. of the receptor. Cell 33, 967–978 (1983)
Cancer Sci. 106, 959–964 (2015) 21. C. Harding, J. Heuser, P. Stahl, Endocytosis and intracellular pro-
4. C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, cessing of transferrin and colloidal gold-transferrin in rat reticulo-
biogenesis and function. Nat. Rev. Immunol. 2, 569–579 (2002) cytes: demonstration of a pathway for receptor shedding. Eur. J.
5. L. Muller, M. Mitsuhashi, P. Simms, W.E. Gooding, T.L. Cell Biol. 35, 256–263 (1984)
Whiteside, Tumor-derived exosomes regulate expression of im- 22. L. Balaj, R. Lessard, L. Dai, Y.J. Cho, S.L. Pomeroy, X.O.
mune function-related genes in human T cell subsets. Sci. Rep. 6, Breakefield, J. Skog, Tumour microvesicles contain
20254 (2016) retrotransposon elements and amplified oncogene sequences.
6. H. Zhao, L. Yang, J. Baddour, A. Achreja, V. Bernard, T. Moss, Nat. Commun. 2, 180 (2011)
J.C. Marini, T. Tudawe, E.G. Seviour, F.A. San Lucas, H. Alvarez, 23. H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O.
S. Gupta, S.N. Maiti, L. Cooper, D. Peehl, P.T. Ram, A. Maitra, D. Lotvall, Exosome-mediated transfer of mRNAs and microRNAs
Nagrath, Tumor microenvironment derived exosomes is a novel mechanism of genetic exchange between cells. Nat. Cell
pleiotropically modulate cancer cell metabolism. elife 5, e10250 Biol. 9, 654–659 (2007)
(2016) 24. C. Admyre, S.M. Johansson, K.R. Qazi, J.J. Filen, R. Lahesmaa,
7. A.S. Azmi, B. Bao, F.H. Sarkar, Exosomes in cancer develop- M. Norman, E.P. Neve, A. Scheynius, S. Gabrielsson, Exosomes
ment, metastasis and drug resistance: a comprehensive review. with immune modulatory features are present in human breast
Cancer Metastasis Rev. 32, 623–642 (2013). https://doi.org/10. milk. J. Immunol. 179, 1969–1978 (2007)
1007/s10555-013-9441-9 25. M.P. Caby, D. Lankar, C. Vincendeau-Scherrer, G. Raposo, C.
8. S.N. Chatterjee, J. Das, Electron microscopic observations on the Bonnerot, Exosomal-like vesicles are present in human blood
excretion of cell-wall material by Vibrio cholerae. J. Gen. plasma. Int. Immunol. 17, 879–887 (2005)
Microbiol. 49, 1–11 (1967) 26. R. Shi, P.Y. Wang, X.Y. Li, J.X. Chen, Y. Li, X.Z. Zhang, C.G.
9. T.N. Ellis, M.J. Kuehn, Virulence and immunomodulatory roles of Zhang, T. Jiang, W.B. Li, W. Ding, S.J. Cheng, Exosomal levels of
bacterial outer membrane vesicles. Microbiol. Mol. Biol. Rev. 74, miRNA-21 from cerebrospinal fluids associated with poor prog-
81–94 (2010) nosis and tumor recurrence of glioma patients. Oncotarget 6,
10. X. Yu, S.L. Harris, A.J. Levine, The regulation of exosome secre- 26971–26981 (2015)
tion: a novel function of the p53 protein. Cancer Res. 66, 4795– 27. M. Gonzalez-Begne, B. Lu, X. Han, F.K. Hagen, A.R. Hand, J.E.
4801 (2006) Melvin, J.R. Yates, Proteomic analysis of human parotid gland
11. A. Lespagnol, D. Duflaut, C. Beekman, L. Blanc, G. Fiucci, J.C. exosomes by multidimensional protein identification technology
Marine, M. Vidal, R. Amson, A. Telerman, Exosome secretion, (MudPIT). J. Proteome Res. 8, 1304–1314 (2009)
including the DNA damage-induced p53-dependent secretory 28. M. Tokuhisa, Y. Ichikawa, N. Kosaka, T. Ochiya, M. Yashiro, K.
pathway, is severely compromised in TSAP6/Steap3-null mice. Hirakawa, T. Kosaka, H. Makino, H. Akiyama, C. Kunisaki, I.
Cell Death Differ. 15, 1723–1733 (2008) Endo, Exosomal miRNAs from peritoneum lavage fluid as poten-
12. C. Thery, M. Ostrowski, E. Segura, Membrane vesicles as tial prognostic biomarkers of peritoneal metastasis in gastric can-
conveyors of immune responses. Nat. Rev. Immunol. 9, cer. PLoS One 10, e0130472 (2015)
581–593 (2009) 29. T. Pisitkun, R.F. Shen, M.A. Knepper, Identification and proteo-
13. W. Li, Y. Hu, T. Jiang, Y. Han, G. Han, J. Chen, X. Li, Rab27A mic profiling of exosomes in human urine. Proc. Natl. Acad. Sci.
regulates exosome secretion from lung adenocarcinoma cells U. S. A. 101, 13368–13373 (2004)
A549: involvement of EPI64. APMIS 122, 1080–1087 (2014) 30. D.D. Taylor, C. Gercel-Taylor, MicroRNA signatures of tumor-
14. I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi, A. De derived exosomes as diagnostic biomarkers of ovarian cancer.
Milito, C. Coscia, E. Iessi, M. Logozzi, A. Molinari, M. Colone, Gynecol. Oncol. 110, 13–21 (2008)
M. Tatti, M. Sargiacomo, S. Fais, Microenvironmental pH is a key 31. C. Thery, S. Amigorena, G. Raposo and A. Clayton, Isolation
factor for exosome traffic in tumor cells. J. Biol. Chem. 284, and characterization of exosomes from cell culture superna-
34211–34222 (2009) tants and biological fluids. Curr. Protoc. Cell Biol.
15. J. Faure, G. Lachenal, M. Court, J. Hirrlinger, C. Chatellard- Chapter 3, Unit 3 22 (2006)
Causse, B. Blot, J. Grange, G. Schoehn, Y. Goldberg, V. Boyer, 32. K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel, F.
F. Kirchhoff, G. Raposo, J. Garin, R. Sadoul, Exosomes are re- Wieland, P. Schwille, B. Brugger, M. Simons, Ceramide triggers
leased by cultured cortical neurones. Mol. Cell. Neurosci. 31, budding of exosome vesicles into multivesicular endosomes.
642–648 (2006) Science 319, 1244–1247 (2008)
16. G. Lachenal, K. Pernet-Gallay, M. Chivet, F.J. Hemming, A. 33. T. Wollert, J.H. Hurley, Molecular mechanism of multivesicular
Belly, G. Bodon, B. Blot, G. Haase, Y. Goldberg, R. Sadoul, body biogenesis by ESCRT complexes. Nature 464, 864–869
Release of exosomes from differentiated neurons and its regula- (2010)
tion by synaptic glutamatergic activity. Mol. Cell. Neurosci. 46, 34. T. Ravid, J.M. Heidinger, P. Gee, E.M. Khan, T. Goldkorn, c-Cbl-
409–418 (2011) mediated ubiquitinylation is required for epidermal growth factor
17. N. Blanchard, D. Lankar, F. Faure, A. Regnault, C. Dumont, G. receptor exit from the early endosomes. J. Biol. Chem. 279,
Raposo, C. Hivroz, TCR activation of human T cells induces the 37153–37162 (2004)
production of exosomes bearing the TCR/CD3/zeta complex. J. 35. L. Duan, Y. Miura, M. Dimri, B. Majumder, I.L. Dodge, A.L.
Immunol. 168, 3235–3241 (2002) Reddi, A. Ghosh, N. Fernandes, P. Zhou, K. Mullane-Robinson,
18. C.T. Roberts Jr., P. Kurre, Vesicle trafficking and RNA transfer N. Rao, S. Donoghue, R.A. Rogers, D. Bowtell, M. Naramura, H.
add complexity and connectivity to cell-cell communication. Gu, V. Band, H. Band, Cbl-mediated ubiquitinylation is required for
Cancer Res. 73, 3200–3205 (2013) lysosomal sorting of epidermal growth factor receptor but is dis-
19. E.G. Trams, C.J. Lauter, N. Salem Jr., U. Heine, Exfoliation of pensable for endocytosis. J. Biol. Chem. 278, 28950–28960 (2003)
membrane ecto-enzymes in the form of micro-vesicles. Biochim. 36. O. Schmidt, D. Teis, The ESCRT machinery. Curr. Biol. 22,
Biophys. Acta 645, 63–70 (1981) R116–R120 (2012)
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 245

37. S. Stuffers, C. Sem Wegner, H. Stenmark, A. Brech, 55. C. Hsu, Y. Morohashi, S. Yoshimura, N. Manrique-Hoyos, S.
Multivesicular endosome biogenesis in the absence of ESCRTs. Jung, M.A. Lauterbach, M. Bakhti, M. Gronborg, W. Mobius, J.
Traffic 10, 925–937 (2009) Rhee, F.A. Barr, M. Simons, Regulation of exosome secretion by
38. T. Kajimoto, T. Okada, S. Miya, L. Zhang, S. Nakamura, Ongoing Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell
activation of sphingosine 1-phosphate receptors mediates maturation Biol. 189, 223–232 (2010)
of exosomal multivesicular endosomes. Nat. Commun. 4, 2712 56. C.A. Thompson, A. Purushothaman, V.C. Ramani, I. Vlodavsky,
(2013) R.D. Sanderson, Heparanase regulates secretion, composition, and
39. D. Perez-Hernandez, C. Gutierrez-Vazquez, I. Jorge, S. Lopez- function of tumor cell-derived exosomes. J. Biol. Chem. 288,
Martin, A. Ursa, F. Sanchez-Madrid, J. Vazquez, M. Yanez-Mo, 10093–10099 (2013)
The intracellular interactome of tetraspanin-enriched microdo- 57. A. Savina, C.M. Fader, M.T. Damiani, M.I. Colombo, Rab11 pro-
mains reveals their function as sorting machineries toward motes docking and fusion of multivesicular bodies in a calcium-
exosomes. J. Biol. Chem. 288, 11649–11661 (2013) dependent manner. Traffic 6, 131–143 (2005)
40. A.V. Vlassov, S. Magdaleno, R. Setterquist, R. Conrad, 58. H.W. King, M.Z. Michael, J.M. Gleadle, Hypoxic enhancement of
Exosomes: current knowledge of their composition, biological exosome release by breast cancer cells. BMC Cancer 12, 1–10
functions, and diagnostic and therapeutic potentials. Biochim. (2012)
Biophys. Acta 1820, 940–948 (2012) 59. A.Z. Ayob, T.S. Ramasamy, Cancer stem cells as key drivers of
41. M. Record, K. Carayon, M. Poirot, S. Silvente-Poirot, Exosomes tumour progression. J. Biomed. Sci. 25, 20 (2018)
as new vesicular lipid transporters involved in cell-cell communi- 60. K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A.
cation and various pathophysiologies. Biochim. Biophys. Acta Guha, J. Rak, Intercellular transfer of the oncogenic receptor
1841, 108–120 (2014) EGFRvIII by microvesicles derived from tumour cells. Nat. Cell
42. F. Coutant, L. Perrin-Cocon, S. Agaugué, T. Delair, P. André, V. Biol. 10, 619–624 (2008)
Lotteau, Mature dendritic cell generation promoted by 61. M.M. Valenzuela, H.R. Ferguson Bennit, A. Gonda, C.J. Diaz
lysophosphatidylcholine. J. Immunol. 169, 1688–1695 (2002) Osterman, A. Hibma, S. Khan, N.R. Wall, Exosomes secreted
43. L. Perrin-Cocon, S. Agaugué, F. Coutant, A. Masurel, S. Bezzine, from human cancer cell lines contain inhibitors of apoptosis
G. Lambeau, P. André, V. Lotteau, Secretory phospholipase A2 (IAP). Cancer Microenviron. 8, 65–73 (2015)
induces dendritic cell maturation. Eur. J. Immunol. 34, 2293–2302 62. E. Donnarumma, D. Fiore, M. Nappa, G. Roscigno, A. Adamo,
(2004) M. Iaboni, V. Russo, A. Affinito, I. Puoti, C. Quintavalle, A.
44. Q. Ge, Y. Zhou, J. Lu, Y. Bai, X. Xie, Z. Lu, miRNA in plasma Rienzo, S. Piscuoglio, R. Thomas, G. Condorelli, Cancer-
exosome is stable under different storage conditions. Molecules associated fibroblasts release exosomal microRNAs that dictate
19, 1568–1575 (2014) an aggressive phenotype in breast cancer. Oncotarget 8, 19592–
19608 (2017)
45. C. Kahlert, R. Kalluri, Exosomes in tumor microenvironment in-
63. A. Ramteke, H. Ting, C. Agarwal, S. Mateen, R. Somasagara, A.
fluence cancer progression and metastasis. J. Mol. Med. 91, 431–
Hussain, M. Graner, B. Frederick, R. Agarwal, G. Deep,
437 (2013)
Exosomes secreted under hypoxia enhance invasiveness and
46. L.A. Mulcahy, R.C. Pink and D.R.F. Carter, Routes and mecha-
stemness of prostate cancer cells by targeting adherens junction
nisms of extracellular vesicle uptake. J. Extracell. Vesicles 3,
molecules. Mol. Carcinog. 54, 554–565 (2015)
https://doi.org/10.3402/jev.v3403.24641 (2014)
64. S. Bao, Q. Wu, S. Sathornsumetee, Y. Hao, Z. Li, A.B.
47. T. Tian, Y.L. Zhu, Y.Y. Zhou, G.F. Liang, Y.Y. Wang, F.H. Hu, Hjelmeland, Q. Shi, R.E. McLendon, D.D. Bigner, J.N. Rich,
Z.D. Xiao, Exosome uptake through clathrin-mediated endocyto- Stem cell-like glioma cells promote tumor angiogenesis through
sis and macropinocytosis and mediating miR-21 delivery. J. Biol. vascular endothelial growth factor. Cancer Res. 66, 7843–7848
Chem. 289, 22258–22267 (2014) (2006)
48. K.J. Svensson, H.C. Christianson, A. Wittrup, E. Bourseau- 65. L. Ricci-Vitiani, R. Pallini, M. Biffoni, M. Todaro, G. Invernici, T.
Guilmain, E. Lindqvist, L.M. Svensson, M. Morgelin, M. Cenci, G. Maira, E.A. Parati, G. Stassi, L.M. Larocca, R. De
Belting, Exosome uptake depends on ERK1/2-heat shock protein Maria, Tumour vascularization via endothelial differentiation of
27 signaling and lipid Raft-mediated endocytosis negatively reg- glioblastoma stem-like cells. Nature 468, 824–828 (2010)
ulated by caveolin-1. J. Biol. Chem. 288, 17713–17724 (2013) 66. P. Carmeliet, R.K. Jain, Angiogenesis in cancer and other diseases.
49. D. Zech, S. Rana, M.W. Büchler, M. Zöller, Tumor-exosomes and Nature 407, 249–257 (2000)
leukocyte activation: an ambivalent crosstalk. Cell Commun. 67. C. Grange, M. Tapparo, F. Collino, L. Vitillo, C. Damasco, M.C.
Signaling 10, 37 (2012) Deregibus, C. Tetta, B. Bussolati, G. Camussi, Microvesicles re-
50. S. Rana, S. Yue, D. Stadel, M. Zoller, Toward tailored exosomes: leased from human renal cancer stem cells stimulate angiogenesis
the exosomal tetraspanin web contributes to target cell selection. and formation of lung premetastatic niche. Cancer Res. 71, 5346–
Int. J. Biochem. Cell Biol. 44, 1574–1584 (2012) 5356 (2011)
51. T.I. Naslund, D. Paquin-Proulx, P.T. Paredes, H. Vallhov, J.K. 68. A. Conigliaro, V. Costa, A. Lo Dico, L. Saieva, S. Buccheri, F.
Sandberg, S. Gabrielsson, Exosomes from breast milk inhibit Dieli, M. Manno, S. Raccosta, C. Mancone, M. Tripodi, G. De
HIV-1 infection of dendritic cells and subsequent viral transfer to Leo, R. Alessandro, CD90+ liver cancer cells modulate endothe-
CD4+ T cells. AIDS 28, 171–180 (2014) lial cell phenotype through the release of exosomes containing
52. J.R. Goldenring, A central role for vesicle trafficking in epithelial H19 lncRNA. Mol. Cancer 14, 155 (2015)
neoplasia: intracellular highways to carcinogenesis. Nat. Rev. 69. E.J. Ekstrom, C. Bergenfelz, V. von Bulow, F. Serifler, E.
Cancer 13, 813–820 (2013) Carlemalm, G. Jonsson, T. Andersson, K. Leandersson,
53. N. Jae, D.G. McEwan, Y. Manavski, R.A. Boon, S. Dimmeler, WNT5A induces release of exosomes containing pro-angiogenic
Rab7a and Rab27b control secretion of endothelial microRNA and immunosuppressive factors from malignant melanoma cells.
through extracellular vesicles. FEBS Lett. 589, 3182–3188 (2015) Mol. Cancer 13, 88 (2014)
54. S.N. Hurwitz, M.M. Conlon, M.A. Rider, N.C. Brownstein, D.G. 70. S.K. Gopal, D.W. Greening, E.G. Hanssen, H.J. Zhu, R.J.
Meckes Jr., Nanoparticle analysis sheds budding insights into ge- Simpson, R.A. Mathias, Oncogenic epithelial cell-derived
netic drivers of extracellular vesicle biogenesis. J. Extracell. exosomes containing Rac1 and PAK2 induce angiogenesis in re-
Vesicles 5, 31295 (2016) cipient endothelial cells. Oncotarget 7, 19709–19722 (2016)
246 V. Sundararajan et al.

71. Y. Liu, F. Luo, B. Wang, H. Li, Y. Xu, X. Liu, L. Shi, X. Lu, W. squamous cell carcinoma cells deliver mir-21 to normoxic cells
Xu, L. Lu, Y. Qin, Q. Xiang, Q. Liu, STAT3-regulated exosomal to elicit a prometastatic phenotype. Cancer Res. 76, 1770–1780
miR-21 promotes angiogenesis and is involved in neoplastic pro- (2016)
cesses of transformed human bronchial epithelial cells. Cancer 87. J. Liao, R. Liu, Y.J. Shi, L.H. Yin, Y.P. Pu, Exosome-shuttling
Lett. 370, 125–135 (2016) microRNA-21 promotes cell migration and invasion-targeting
72. Y.K. Chan, H. Zhang, P. Liu, S.W. Tsao, M.L. Lung, N.K. Mak, R. PDCD4 in esophageal cancer. Int. J. Oncol. 48, 2567–2579 (2016)
Ngok-Shun Wong, P. Ying-Kit Yue, Proteomic analysis of 88. M. Yang, J. Chen, F. Su, B. Yu, F. Su, L. Lin, Y. Liu, J.-D. Huang,
exosomes from nasopharyngeal carcinoma cell identifies intercellu- E. Song, Microvesicles secreted by macrophages shuttle invasion-
lar transfer of angiogenic proteins. Int. J. Cancer 137, 1830–1841 potentiating microRNAs into breast cancer cells. Mol. Cancer 10,
(2015) 1–13 (2011)
73. K. Pakravan, S. Babashah, M. Sadeghizadeh, S.J. Mowla, M. 89. X.L. Bai, Q. Zhang, L.Y. Ye, F. Liang, X. Sun, Y. Chen, Q.D. Hu,
Mossahebi-Mohammadi, F. Ataei, N. Dana, M. Javan, Q.H. Fu, W. Su, Z. Chen, Z.P. Zhuang, T.B. Liang, Myocyte
MicroRNA-100 shuttled by mesenchymal stem cell-derived enhancer factor 2C regulation of hepatocellular carcinoma via
exosomes suppresses in vitro angiogenesis through modulating vascular endothelial growth factor and Wnt/beta-catenin signaling.
the mTOR/HIF-1alpha/VEGF signaling axis in breast cancer cells. Oncogene 34, 4089–4097 (2015)
Cell. Oncol. 40, 457–470 (2017) 90. M.Y. Fong, W. Zhou, L. Liu, A.Y. Alontaga, M. Chandra, J.
74. H. Tadokoro, T. Umezu, K. Ohyashiki, T. Hirano, J.H. Ohyashiki, Ashby, A. Chow, S.T. O'Connor, S. Li, A.R. Chin, G. Somlo,
Exosomes derived from hypoxic leukemia cells enhance tube for- M. Palomares, Z. Li, J.R. Tremblay, A. Tsuyada, G. Sun, M.A.
mation in endothelial cells. J. Biol. Chem. 288, 34343–34351 Reid, X. Wu, P. Swiderski, X. Ren, Y. Shi, M. Kong, W. Zhong, Y.
(2013) Chen, S.E. Wang, Breast-cancer-secreted miR-122 reprograms
75. T. Umezu, H. Tadokoro, K. Azuma, S. Yoshizawa, K. Ohyashiki, glucose metabolism in premetastatic niche to promote metastasis.
J.H. Ohyashiki, Exosomal miR-135b shed from hypoxic multiple Nat. Cell Biol. 17, 183–194 (2015)
myeloma cells enhances angiogenesis by targeting factor- 91. M.M. Gottesman, Mechanisms of cancer drug resistance. Annu.
inhibiting HIF-1. Blood 124, 3748–3757 (2014) Rev. Med. 53, 615–627 (2002)
76. R. Kalluri, R.A. Weinberg, The basics of epithelial-mesenchymal
92. W.X. Chen, X.M. Liu, M.M. Lv, L. Chen, J.H. Zhao, S.L. Zhong,
transition. J. Clin. Invest. 119, 1420–1428 (2009)
M.H. Ji, Q. Hu, Z. Luo, J.Z. Wu, J.H. Tang, Exosomes from drug-
77. S. Shi, Q. Zhang, Y. Xia, B. You, Y. Shan, L. Bao, L. Li, Y. You, Z.
resistant breast cancer cells transmit chemoresistance by a hori-
Gu, Mesenchymal stem cell-derived exosomes facilitate nasopha-
zontal transfer of microRNAs. PLoS One 9, e95240 (2014)
ryngeal carcinoma progression. Am. J. Cancer Res. 6, 459–472
93. M.M. Lv, X.Y. Zhu, W.X. Chen, S.L. Zhong, Q. Hu, T.F. Ma, J.
(2016)
Zhang, L. Chen, J.H. Tang, J.H. Zhao, Exosomes mediate drug
78. C.A. Franzen, R.H. Blackwell, V. Todorovic, K.A. Greco, K.E.
resistance transfer in MCF-7 breast cancer cells and a probable
Foreman, R.C. Flanigan, P.C. Kuo, G.N. Gupta, Urothelial cells
mechanism is delivery of P-glycoprotein. Tumour Biol. 35,
undergo epithelial-to-mesenchymal transition after exposure to
10773–10779 (2014)
muscle invasive bladder cancer exosomes. Oncogene 4, e163
(2015) 94. C.L. Au Yeung, N.N. Co, T. Tsuruga, T.L. Yeung, S.Y. Kwan, C.S.
Leung, Y. Li, E.S. Lu, K. Kwan, K.K. Wong, R. Schmandt, K.H.
79. M. Aga, G.L. Bentz, S. Raffa, M.R. Torrisi, S. Kondo, N. Wakisaka,
Lu, S.C. Mok, Exosomal transfer of stroma-derived miR21 con-
T. Yoshizaki, J.S. Pagano, J. Shackelford, Exosomal HIF1alpha
fers paclitaxel resistance in ovarian cancer cells through targeting
supports invasive potential of nasopharyngeal carcinoma-
APAF1. Nat. Commun. 7, 11150 (2016)
associated LMP1-positive exosomes. Oncogene 33, 4613–4622
(2014) 95. Y. Hu, C. Yan, L. Mu, K. Huang, X. Li, D. Tao, Y. Wu, J. Qin,
80. W. Qin, Y. Tsukasaki, S. Dasgupta, N. Mukhopadhyay, M. Ikebe, Fibroblast-derived exosomes contribute to chemoresistance
E.R. Sauter, Exosomes in human breast milk promote emt. Clin. through priming cancer stem cells in colorectal cancer. PLoS
Cancer Res. 22, 4517–4524 (2016) One 10, e0125625 (2015)
81. J. Zhang, L. Ma, MicroRNA control of epithelial-mesenchymal 96. R. Ji, B. Zhang, X. Zhang, J. Xue, X. Yuan, Y. Yan, M. Wang, W.
transition and metastasis. Cancer Metastasis Rev. 31, 653–662 Zhu, H. Qian, W. Xu, Exosomes derived from human mesenchy-
(2012) mal stem cells confer drug resistance in gastric cancer. Cell Cycle
82. D. Xiao, S. Barry, D. Kmetz, M. Egger, J. Pan, S.N. Rai, J. Qu, 14, 2473–2483 (2015)
K.M. McMasters, H. Hao, Melanoma cell-derived exosomes pro- 97. T. Aung, B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann, M.
mote epithelial-mesenchymal transition in primary melanocytes Lahmann, T. Weinhage, K. Menck, T. Hupfeld, R. Koch, L.
through paracrine/autocrine signaling in the tumor microenviron- Trumper, G.G. Wulf, Exosomal evasion of humoral immunother-
ment. Cancer Lett. 376, 318–327 (2016) apy in aggressive B-cell lymphoma modulated by ATP-binding
83. I.J. Fidler, The pathogenesis of cancer metastasis: the 'seed and cassette transporter A3. Proc. Natl. Acad. Sci. U. S. A. 108,
soil' hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003) 15336–15341 (2011)
84. M. Rodriguez, J. Silva, A. Herrera, M. Herrera, C. Pena, P. Martin, 98. V. Ciravolo, V. Huber, G.C. Ghedini, E. Venturelli, F. Bianchi, M.
B. Gil-Calderon, M.J. Larriba, M.J. Coronado, B. Soldevilla, V.S. Campiglio, D. Morelli, A. Villa, P. Della Mina, S. Menard, P.
Turrion, M. Provencio, A. Sanchez, F. Bonilla, V. Garcia- Filipazzi, L. Rivoltini, E. Tagliabue, S.M. Pupa, Potential role of
Barberan, Exosomes enriched in stemness/metastatic-related HER2-overexpressing exosomes in countering trastuzumab-based
mRNAS promote oncogenic potential in breast cancer. therapy. J. Cell. Physiol. 227, 658–667 (2012)
Oncotarget 6, 40575–40587 (2015) 99. R. Safaei, B.J. Larson, T.C. Cheng, M.A. Gibson, S. Otani, W.
85. T. Arita, D. Ichikawa, H. Konishi, S. Komatsu, A. Shiozaki, S. Naerdemann, S.B. Howell, Abnormal lysosomal trafficking and
Ogino, Y. Fujita, H. Hiramoto, J. Hamada, K. Shoda, T. Kosuga, enhanced exosomal export of cisplatin in drug-resistant human
H. Fujiwara, K. Okamoto, E. Otsuji, Tumor exosome-mediated ovarian carcinoma cells. Mol. Cancer Ther. 4, 1595–1604 (2005)
promotion of adhesion to mesothelial cells in gastric cancer cells. 100. S. Loewer, M.N. Cabili, M. Guttman, Y.H. Loh, K. Thomas, I.H.
Oncotarget 7, 56855–56863 (2016) Park, M. Garber, M. Curran, T. Onder, S. Agarwal, P.D. Manos, S.
86. L. Li, C. Li, S. Wang, Z. Wang, J. Jiang, W. Wang, X. Li, J. Chen, Datta, E.S. Lander, T.M. Schlaeger, G.Q. Daley, J.L. Rinn, Large
K. Liu, C. Li, G. Zhu, Exosomes derived from hypoxic oral intergenic non-coding RNA-RoR modulates reprogramming of
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 247

human induced pluripotent stem cells. Nat. Genet. 42, 1113–1117 release from human prostate cancer cells. Cancer Res. 68, 7864–
(2010) 7871 (2008)
101. Y. Pan, C. Li, J. Chen, K. Zhang, X. Chu, R. Wang, L. Chen, The 119. K. Weiner-Gorzel, E. Dempsey, M. Milewska, A. McGoldrick, V.
emerging roles of long noncoding rna ror (lincrna-ror) and its Toh, A. Walsh, S. Lindsay, L. Gubbins, A. Cannon, D. Sharpe, J.
possible mechanisms in human cancers. Cell. Physiol. Biochem. O'Sullivan, M. Murphy, S.F. Madden, M. Kell, A. McCann, F.
40, 219–229 (2016) Furlong, Overexpression of the microRNA miR-433 promotes
102. K. Takahashi, I.K. Yan, T. Kogure, H. Haga, T. Patel, Extracellular resistance to paclitaxel through the induction of cellular senes-
vesicle-mediated transfer of long non-coding RNA ROR modu- cence in ovarian cancer cells. Cancer Med. 4, 745–758 (2015)
lates chemosensitivity in human hepatocellular cancer. FEBS 120. F. Furlong, P. Fitzpatrick, S. O'Toole, S. Phelan, B. McGrogan, A.
Open Bio. 4, 458–467 (2014) Maguire, A. O'Grady, M. Gallagher, M. Prencipe, A. McGoldrick,
103. J. Fan, Y. Xing, X. Wen, R. Jia, H. Ni, J. He, X. Ding, H. Pan, G. P. McGettigan, D. Brennan, O. Sheils, C. Martin, E. W. Kay, J.
Qian, S. Ge, A.R. Hoffman, H. Zhang, X. Fan, Long non-coding O'Leary, A. McCann, Low MAD2 expression levels associate with
RNA ROR decoys gene-specific histone methylation to promote reduced progression-free survival in patients with high-grade serous
tumorigenesis. Genome Biol. 16, 139 (2015) epithelial ovarian cancer. J. Pathol. 226, 746–755 (2012)
104. T.H. Cheung, T.A. Rando, Molecular regulation of stem cell qui- 121. B.W. van Balkom, O.G. de Jong, M. Smits, J. Brummelman, K.
escence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013) den Ouden, P.M. de Bree, M.A. van Eijndhoven, D.M. Pegtel, W.
105. P.K. Lim, S.A. Bliss, S.A. Patel, M. Taborga, M.A. Dave, L.A. Stoorvogel, T. Wurdinger, M.C. Verhaar, Endothelial cells require
Gregory, S.J. Greco, M. Bryan, P.S. Patel, P. Rameshwar, Gap miR-214 to secrete exosomes that suppress senescence and induce
junction-mediated import of microRNA from bone marrow stro- angiogenesis in human and mouse endothelial cells. Blood 121,
mal cells can elicit cell cycle quiescence in breast cancer cells. 3997–4006 (2013)
Cancer Res. 71, 1550–1560 (2011) 122. S. Baroni, S. Romero-Cordoba, I. Plantamura, M. Dugo, E.
106. M. Ono, N. Kosaka, N. Tominaga, Y. Yoshioka, F. Takeshita, R.-u. D'Ippolito, A. Cataldo, G. Cosentino, V. Angeloni, A. Rossini,
Takahashi, M. Yoshida, H. Tsuda, K. Tamura, T. Ochiya, M.G. Daidone, M.V. Iorio, Exosome-mediated delivery of miR-
Exosomes from bone marrow mesenchymal stem cells contain a 9 induces cancer-associated fibroblast-like properties in human
microRNA that promotes dormancy in metastatic breast cancer breast fibroblasts. Cell Death Dis. 7, e2312 (2016)
cells. Sci. Signal. 7, ra63 (2014) 123. A. Gutkin, O. Uziel, E. Beery, J. Nordenberg, M. Pinchasi, H.
107. S.A. Bliss, G. Sinha, O. Sandiford, L. Williams, D.J. Engelberth, K. Goldvaser, S. Henick, M. Goldberg, M. Lahav, Tumor cells de-
Guiro, L.L. Isenalumhe, S.J. Greco, S. Ayer, M. Bryan, R. Kumar, rived exosomes contain hTERT mRNA and transform nonmalig-
N. Ponzio, P. Rameshwar, Mesenchymal stem cell-derived nant fibroblasts into telomerase positive cells. Oncotarget 7,
exosomes stimulates cycling quiescence and early breast cancer 59173–59188 (2016)
dormancy in bone marrow. Cancer Res. 76, 5832–5844 (2016) 124. J. Gu, H. Qian, L. Shen, X. Zhang, W. Zhu, L. Huang, Y. Yan, F.
108. M. Collado, M.A. Blasco, M. Serrano, Cellular senescence in Mao, C. Zhao, Y. Shi, W. Xu, Gastric cancer exosomes trigger
cancer and aging. Cell 130, 223–233 (2007) differentiation of umbilical cord derived mesenchymal stem cells
109. P. Kahlem, B. Dorken, C.A. Schmitt, Cellular senescence in can- to carcinoma-associated fibroblasts through TGF-beta/Smad path-
cer treatment: friend or foe? J. Clin. Invest. 113, 169–174 (2004) way. PLoS One 7, e52465 (2012)
110. H.D. Skinner, V.C. Sandulache, T.J. Ow, R.E. Meyn, J.S. Yordy, 125. J. Webber, R. Steadman, M.D. Mason, Z. Tabi, A. Clayton, Cancer
B.M. Beadle, A.L. Fitzgerald, U. Giri, K.K. Ang, J.N. Myers, exosomes trigger fibroblast to myofibroblast differentiation.
TP53 disruptive mutations lead to head and neck cancer treatment Cancer Res. 70, 9621–9630 (2010)
failure through inhibition of radiation-induced senescence. Clin. 126. A. Orimo, Y. Tomioka, Y. Shimizu, M. Sato, S. Oigawa, K.
Cancer Res. 18, 290–300 (2012) Kamata, Y. Nogi, S. Inoue, M. Takahashi, T. Hata, M.
111. B. Jonchere, A. Vetillard, B. Toutain, D. Lam, A.C. Bernard, C. Muramatsu, Cancer-associated myofibroblasts possess various
Henry, S. De Carne Trecesson, E. Gamelin, P. Juin, C. Guette, O. factors to promote endometrial tumor progression. Clin. Cancer
Coqueret, Irinotecan treatment and senescence failure promote the Res. 7, 3097–3105 (2001)
emergence of more transformed and invasive cells that depend on 127. L.M. Sobral, A. Bufalino, M.A. Lopes, E. Graner, T. Salo, R.D.
anti-apoptotic Mcl-1. Oncotarget 6, 409–426 (2015) Coletta, Myofibroblasts in the stroma of oral cancer promote tumor-
112. A.L.C. Ong, T.S. Ramasamy, Role of sirtuin1-p53 regulatory axis igenesis via secretion of activin A. Oral Oncol. 47, 840–846 (2011)
in aging, cancer and cellular reprogramming. Ageing Res. Rev. 43, 128. S. Vong, R. Kalluri, The role of stromal myofibroblast and
64–80 (2018) extracellular matrix in tumor angiogenesis. Genes Cancer 2,
113. X. Yu, T. Riley, A.J. Levine, The regulation of the endosomal 1139–1145 (2011)
compartment by p53 the tumor suppressor gene. FEBS J. 276, 129. C. Corrado, L. Saieva, S. Raimondo, A. Santoro, G. De Leo, R.
2201–2212 (2009) Alessandro, Chronic myelogenous leukaemia exosomes modulate
114. Y. Sun, W. Zheng, Z. Guo, Q. Ju, L. Zhu, J. Gao, L. Zhou, F. Liu, bone marrow microenvironment through activation of epidermal
Y. Xu, Q. Zhan, Z. Zhou, W. Sun, X. Zhao, A novel TP53 pathway growth factor receptor. J. Cell. Mol. Med. 20, 1829–1839 (2016)
influences the HGS-mediated exosome formation in colorectal 130. L. Wu, X. Zhang, B. Zhang, H. Shi, X. Yuan, Y. Sun, Z. Pan, H.
cancer. Sci. Rep. 6, 28083 (2016) Qian, W. Xu, Exosomes derived from gastric cancer cells activate
115. N. Malaquin, A. Martinez, F. Rodier, Keeping the senescence NF-kappaB pathway in macrophages to promote cancer progres-
secretome under control: Molecular reins on the senescence- sion. Tumour Biol. 37, 12169–12180 (2016)
associated secretory phenotype. Exp. Gerontol. 82, 39–49 (2016) 131. M. Egeblad, E.S. Nakasone, Z. Werb, Tumors as organs: complex
116. J.P. Coppe, K. Kauser, J. Campisi, C.M. Beausejour, Secretion of tissues that interface with the entire organism. Dev. Cell 18, 884–
vascular endothelial growth factor by primary human fibroblasts at 901 (2010)
senescence. J. Biol. Chem. 281, 29568–29574 (2006) 132. L.R. Languino, A. Singh, M. Prisco, G.J. Inman, A. Luginbuhl,
117. X. Sun, M. Vale, E. Leung, J.R. Kanwar, R. Gupta, G.W. J.M. Curry, A.P. South, Exosome-mediated transfer from the tu-
Krissansen, Mouse B7-H3 induces antitumor immunity. Gene mor microenvironment increases TGFbeta signaling in squamous
Ther. 10, 1728–1734 (2003) cell carcinoma. Am. J. Transl. Res. 8, 2432–2437 (2016)
118. B.D. Lehmann, M.S. Paine, A.M. Brooks, J.A. McCubrey, R.H. 133. M.C. Boelens, T.J. Wu, B.Y. Nabet, B. Xu, Y. Qiu, T. Yoon, D.J.
Renegar, R. Wang, D.M. Terrian, Senescence-associated exosome Azzam, C. Twyman-Saint Victor, B.Z. Wiemann, H. Ishwaran,
248 V. Sundararajan et al.

P.J. Ter Brugge, J. Jonkers, J. Slingerland, A.J. Minn, Exosome 148. A. De Boeck, P. Pauwels, K. Hensen, J.L. Rummens, W.
transfer from stromal to breast cancer cells regulates therapy resis- Westbroek, A. Hendrix, D. Maynard, H. Denys, K. Lambein, G.
tance pathways. Cell 159, 499–513 (2014) Braems, C. Gespach, M. Bracke, O. De Wever, Bone marrow-
134. D.J. Prockop, Marrow stromal cells as stem cells for derived mesenchymal stem cells promote colorectal cancer pro-
nonhematopoietic tissues. Science 276, 71–74 (1997) gression through paracrine neuregulin 1/HER3 signalling. Gut 62,
135. G. Lazennec, C. Jorgensen, Concise Review: Adult multipotent 550–560 (2013)
stromal cells and cancer: risk or benefit? Stem Cells 26, 1387– 149. Y. Huang, P. Yu, W. Li, G. Ren, A.I. Roberts, W. Cao, X. Zhang, J.
1394 (2008) Su, X. Chen, Q. Chen, P. Shou, C. Xu, L. Du, L. Lin, N. Xie, L.
136. B. Cousin, E. Ravet, S. Poglio, F. De Toni, M. Bertuzzi, H. Lulka, I. Zhang, Y. Wang, Y. Shi, p53 regulates mesenchymal stem cell-
Touil, M. Andre, J.L. Grolleau, J.M. Peron, J.P. Chavoin, P. Bourin, mediated tumor suppression in a tumor microenvironment through
L. Penicaud, L. Casteilla, L. Buscail, P. Cordelier, Adult stromal immune modulation. Oncogene 33, 3830–3838 (2014)
cells derived from human adipose tissue provoke pancreatic cancer 150. K. McLean, Y. Gong, Y. Choi, N. Deng, K. Yang, S. Bai, L.
cell death both in vitro and in vivo. PLoS One 4, e6278 (2009) Cabrera, E. Keller, L. McCauley, K.R. Cho, R.J. Buckanovich,
137. L. Qiao, Z.L. Xu, T.J. Zhao, L.H. Ye, X.D. Zhang, Dkk-1 secreted Human ovarian carcinoma-associated mesenchymal stem cells
by mesenchymal stem cells inhibits growth of breast cancer cells regulate cancer stem cells and tumorigenesis via altered BMP
via depression of Wnt signalling. Cancer Lett. 269, 67–77 (2008) production. J. Clin. Invest. 121, 3206–3219 (2011)
138. Y. Yulyana, I.A. Ho, K.C. Sia, J.P. Newman, X.Y. Toh, B.B. Endaya, 151. F. Vianello, F. Villanova, V. Tisato, S. Lymperi, K.-K. Ho, A.R.
J.K. Chan, M. Gnecchi, H. Huynh, A.Y. Chung, K.H. Lim, H.S. Gomes, D. Marin, D. Bonnet, J. Apperley, E.W.F. Lam, F. Dazzi,
Leong, N.G. Iyer, K.M. Hui, P.Y. Lam, Paracrine factors of human Bone marrow mesenchymal stromal cells non-selectively protect
fetal MSCs inhibit liver cancer growth through reduced activation of chronic myeloid leukemia cells from imatinib-induced apoptosis via
IGF-1R/PI3K/Akt signaling. Mol. Ther. 23, 746–756 (2015) the CXCR4/CXCL12 axis. Haematologica 95, 1081–1089 (2010)
139. O. Attar-Schneider, V. Zismanov, L. Drucker, M. Gottfried, 152. L.Y. Lin, L.M. Du, K. Cao, Y. Huang, P.F. Yu, L.Y. Zhang, F.Y. Li,
Secretome of human bone marrow mesenchymal stem cells: an Y. Wang, Y.F. Shi, Tumour cell-derived exosomes endow mesen-
emerging player in lung cancer progression and mechanisms of chymal stromal cells with tumour-promotion capabilities.
translation initiation. Tumor Biol. 37, 4755–4765 (2016) Oncogene 35, 6038–6042 (2016)
140. J.K. Lee, S.R. Park, B.K. Jung, Y.K. Jeon, Y.S. Lee, M.K. Kim, Y.G. 153. X. Song, Y. Ding, G. Liu, X. Yang, R. Zhao, Y. Zhang, X. Zhao,
Kim, J.Y. Jang, C.W. Kim, Exosomes derived from mesenchymal G.J. Anderson, G. Nie, Cancer Cell-derived exosomes induce
stem cells suppress angiogenesis by down-regulating VEGF expres- mitogen-activated protein kinase-dependent monocyte survival
sion in breast cancer cells. PLoS One 8, e84256 (2013) by transport of functional receptor tyrosine kinases. J. Biol.
141. A.Y. Khakoo, S. Pati, S.A. Anderson, W. Reid, M.F. Elshal, I.I. Chem. 291, 8453–8464 (2016)
Rovira, A.T. Nguyen, D. Malide, C.A. Combs, G. Hall, J. Zhang, 154. J. Choi, J. Gyamfi, H. Jang and J.S. Koo, The role of tumor-
M. Raffeld, T.B. Rogers, W. Stetler-Stevenson, J.A. Frank, M. associated macrophage in breast cancer biology. Histol.
Reitz, T. Finkel, Human mesenchymal stem cells exert potent Histopathol. 33, 133–145 (2018)
antitumorigenic effects in a model of Kaposi's sarcoma. J. Exp. 155. F. Leonard, L.T. Curtis, M.J. Ware, T. Nosrat, X. Liu, K. Yokoi,
Med. 203, 1235–1247 (2006) H.B. Frieboes, B. Godin, Macrophage polarization contributes to
142. J.F. Ji, B.P. He, S.T. Dheen, S.S.W. Tay, Interactions of the anti-tumoral efficacy of mesoporous nanovectors loaded with
chemokines and chemokine receptors mediate the migration of albumin-bound paclitaxel. Front. Immunol. 8, 693 (2017)
mesenchymal stem cells to the impaired site in the brain after 156. Z. Chen, X. Feng, C.J. Herting, V. Alvarez Garcia, K. Nie, W.W.
hypoglossal nerve injury. Stem Cells 22, 415–427 (2004) Pong, R. Rasmussen, B. Dwivedi, S. Seby, S.A. Wolf, D.H.
143. B.M. Beckermann, G. Kallifatidis, A. Groth, D. Frommhold, A. Gutmann, D. Hambardzumyan, Cellular and molecular identity
Apel, J. Mattern, A.V. Salnikov, G. Moldenhauer, W. Wagner, A. of tumor-associated macrophages in glioblastoma. Cancer Res.
Diehlmann, R. Saffrich, M. Schubert, A.D. Ho, N. Giese, M.W. 77, 2266–2278 (2017)
Buchler, H. Friess, P. Buchler, I. Herr, VEGF expression by mes- 157. M. Yin, X. Li, S. Tan, H.J. Zhou, W. Ji, S. Bellone, X. Xu, H.
enchymal stem cells contributes to angiogenesis in pancreatic car- Zhang, A.D. Santin, G. Lou, W. Min, Tumor-associated macro-
cinoma. Br. J. Cancer 99, 622–631 (2008) phages drive spheroid formation during early transcoelomic me-
144. A. Schmidt, D. Ladage, T. Schinköthe, U. Klausmann, C. Ulrichs, tastasis of ovarian cancer. J. Clin. Invest. 126, 4157–4173 (2016)
F.J. Klinz, K. Brixius, S. Arnhold, B. Desai, U. Mehlhorn, R.H.G. 158. H. Shinohara, Y. Kuranaga, M. Kumazaki, N. Sugito, Y.
Schwinger, P. Staib, K. Addicks, W. Bloch, Basic fibroblast Yoshikawa, T. Takai, K. Taniguchi, Y. Ito, Y. Akao, Regulated
growth factor controls migration in human mesenchymal stem polarization of tumor-associated macrophages by mir-145 via co-
cells. Stem Cells 24, 1750–1758 (2006) lorectal cancer–derived extracellular vesicles. J. Immunol. 199,
145. C. Ke, J. Chen, Y. Guo, Z.W. Chen, J. Cai, Migration mechanism 1505–1515 (2017)
of mesenchymal stem cells studied by QD/NSOM. Biochim. 159. J. Wang, K. De Veirman, S. Faict, M.A. Frassanito, D. Ribatti, A.
Biophys. Acta Biomembr. 1848, 859–868 (2015) Vacca, E. Menu, Multiple myeloma exosomes establish a
146. G. Ren, X. Zhao, Y. Wang, X. Zhang, X. Chen, C. Xu, Z.R. Yuan, favourable bone marrow microenvironment with enhanced angio-
A.I. Roberts, L. Zhang, B. Zheng, T. Wen, Y. Han, A.B. Rabson, genesis and immunosuppression. J. Pathol. 239, 162–173 (2016)
J.A. Tischfield, C. Shao, Y. Shi, CCR2-dependent recruitment of 160. U. Putz, J. Howitt, A. Doan, C.-P. Goh, L.-H. Low, J. Silke, S.-S.
macrophages by tumor-educated mesenchymal stromal cells pro- Tan, The tumor suppressor pten is exported in exosomes and has
motes tumor development and is mimicked by TNFalpha. Cell phosphatase activity in recipient cells. Sci. Signal. 5, ra70 (2012)
Stem Cell 11, 812–824 (2012) 161. A.M.M.T. Reza, Y.-J. Choi, H. Yasuda, J.-H. Kim, Human adipose
147. B.G. Cuiffo, A. Campagne, G.W. Bell, A. Lembo, F. Orso, E.C. Lien, mesenchymal stem cell-derived exosomal-miRNAs are critical
M.K. Bhasin, M. Raimo, S.E. Hanson, A. Marusyk, D. El-Ashry, P. factors for inducing anti-proliferation signalling to A2780 and
Hematti, K. Polyak, F. Mechta-Grigoriou, O. Mariani, S. Volinia, A. SKOV-3 ovarian cancer cells. Sci. Rep. 6, 38498 (2016)
Vincent-Salomon, D. Taverna, A.E. Karnoub, MSC-regulated 162. S.F. Ko, H.K. Yip, Y.Y. Zhen, C.C. Lee, C.C. Lee, C.C. Huang, S.H.
microRNAs converge on the transcription factor FOXP2 and promote Ng, J.W. Lin, Adipose-derived mesenchymal stem cell exosomes
breast cancer metastasis. Cell Stem Cell 15, 762–774 (2014) suppress hepatocellular carcinoma growth in a rat model: apparent
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 249

diffusion coefficient, natural killer t-cell responses, and histopatho- Dasgupta, Mitochondrial reprogramming regulates breast cancer
logical features. Stem Cells Int. 2015, 853506 (2015) progression. Clin. Cancer Res. 22, 3348–3360 (2016)
163. F. Alcayaga-Miranda, P.L. Gonzalez, A. Lopez-Verrilli, M. Varas- 175. M.J. Donovan, M. Noerholm, S. Bentink, S. Belzer, J. Skog, V.
Godoy, C. Aguila-Diaz, L. Contreras, M. Khoury, Prostate tumor- O'Neill, J.S. Cochran, G.A. Brown, A molecular signature of
induced angiogenesis is blocked by exosomes derived from men- PCA3 and ERG exosomal RNA from non-DRE urine is predictive
strual stem cells through the inhibition of reactive oxygen species. of initial prostate biopsy result. Prostate Cancer Prostatic Dis. 18,
Oncotarget 7, 44462–44477 (2016) 370–375 (2015)
164. H.D. Lee, B.H. Koo, Y.H. Kim, O.H. Jeon, D.S. Kim, Exosome 176. M. He, H. Qin, T.C. Poon, S.C. Sze, X. Ding, N.N. Co, S.M. Ngai,
release of ADAM15 and the functional implications of human T.F. Chan, N. Wong, Hepatocellular carcinoma-derived exosomes
macrophage-derived ADAM15 exosomes. FASEB J. 26, 3084– promote motility of immortalized hepatocyte through transfer of on-
3095 (2012) cogenic proteins and RNAs. Carcinogenesis 36, 1008–1018 (2015)
165. B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. 177. G.K. Joshi, S. Deitz-McElyea, T. Liyanage, K. Lawrence, S. Mali,
Thakur, A. Becker, A. Hoshino, M.T. Mark, H. Molina, J. Xiang, R. Sardar, M. Korc, Label-free nanoplasmonic-based short noncod-
T. Zhang, T.M. Theilen, G. Garcia-Santos, C. Williams, Y. Ararso, ing rna sensing at attomolar concentrations allows for quantitative
Y. Huang, G. Rodrigues, T.L. Shen, K.J. Labori, I.M. Lothe, E.H. and highly specific assay of microrna-10b in biological fluids and
Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. Grandgenett, circulating exosomes. ACS Nano 9, 11075–11089 (2015)
M.A. Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. 178. L. Manterola, E. Guruceaga, J. Gallego Perez-Larraya, M.
Jarnagin, R.E. Schwartz, I. Matei, H. Peinado, B.Z. Stanger, J. Gonzalez-Huarriz, P. Jauregui, S. Tejada, R. Diez-Valle, V.
Bromberg, D. Lyden, Pancreatic cancer exosomes initiate pre- Segura, N. Sampron, C. Barrena, I. Ruiz, A. Agirre, A. Ayuso,
metastatic niche formation in the liver. Nat. Cell Biol. 17, 816– J. Rodriguez, A. Gonzalez, E. Xipell, A. Matheu, A. Lopez de
826 (2015) Munain, T. Tunon, I. Zazpe, J. Garcia-Foncillas, S. Paris, J.Y.
166. J. Sceneay, M.J. Smyth, A. Moller, The pre-metastatic niche: find- Delattre, M.M. Alonso, A small noncoding RNA signature found
ing common ground. Cancer Metastasis Rev. 32, 449–464 (2013) in exosomes of GBM patient serum as a diagnostic tool. Neuro-
167. H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, Oncology 16, 520–527 (2014)
G. Moreno-Bueno, M. Hergueta-Redondo, C. Williams, G. 179. J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, W.T.
Garcia-Santos, C. Ghajar, A. Nitadori-Hoshino, C. Hoffman, K. Curry, B.S. Carter, A.M. Krichevsky, X.O. Breakefield,
Badal, B.A. Garcia, M.K. Callahan, J. Yuan, V.R. Martins, J. Glioblastoma microvesicles transport RNA and proteins that pro-
Skog, R.N. Kaplan, M.S. Brady, J.D. Wolchok, P.B. Chapman, mote tumour growth and provide diagnostic biomarkers. Nat. Cell
Y. Kang, J. Bromberg, D. Lyden, Melanoma exosomes educate Biol. 10, 1470–1476 (2008)
bone marrow progenitor cells toward a pro-metastatic phenotype
180. P. Kharaziha, D. Chioureas, D. Rutishauser, G. Baltatzis, L.
through MET. Nat. Med. 18, 883–891 (2012)
Lennartsson, P. Fonseca, A. Azimi, K. Hultenby, R. Zubarev, A.
168. T. Jung, D. Castellana, P. Klingbeil, I. Cuesta Hernandez, M.
Ullen, J. Yachnin, S. Nilsson, T. Panaretakis, Molecular profiling of
Vitacolonna, D.J. Orlicky, S.R. Roffler, P. Brodt, M. Zoller,
prostate cancer derived exosomes may reveal a predictive signature
CD44v6 dependence of premetastatic niche preparation by
for response to docetaxel. Oncotarget 6, 21740–21754 (2015)
exosomes. Neoplasia 11, 1093–1105 (2009)
169. J.L. Hood, R.S. San and S.A. Wickline, Exosomes released by 181. K. Kawakami, Y. Fujita, T. Kato, K. Mizutani, K. Kameyama, H.
melanoma cells prepare sentinel lymph nodes for tumor metasta- Tsumoto, Y. Miura, T. Deguchi, M. Ito, Integrin beta4 and vinculin
sis. Cancer Res. 71, 3792–3801 (2011) contained in exosomes are potential markers for progression of
170. C.A. Sanchez, E.I. Andahur, R. Valenzuela, E.A. Castellon, J.A. prostate cancer associated with taxane-resistance. Int. J. Oncol.
Fulla, C.G. Ramos, J.C. Trivino, Exosomes from bulk and stem 47, 384–390 (2015)
cells from human prostate cancer have a differential microRNA 182. Y.Y. Yeh, H.G. Ozer, A.M. Lehman, K. Maddocks, L. Yu, A.J.
content that contributes cooperatively over local and pre- Johnson, J.C. Byrd, Characterization of CLL exosomes reveals a
metastatic niche. Oncotarget 7, 3993–4008 (2016) distinct microRNA signature and enhanced secretion by activation
171. A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. of BCR signaling. Blood 125, 3297–3305 (2015)
Hashimoto, M. Tesic Mark, H. Molina, S. Kohsaka, A. Di 183. J. Silva, V. Garcia, M. Rodriguez, M. Compte, E. Cisneros, P.
Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop, K. Veguillas, J.M. Garcia, G. Dominguez, Y. Campos-Martin, J.
Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, Cuevas, C. Pena, M. Herrera, R. Diaz, N. Mohammed, F. Bonilla,
T. Zhang, H. Zhang, J. Hernandez, J.M. Weiss, V.D. Dumont- Analysis of exosome release and its prognostic value in human
Cole, K. Kramer, L.H. Wexler, A. Narendran, G.K. Schwartz, colorectal cancer. Genes Chromosom. Cancer 51, 409–418 (2012)
J.H. Healey, P. Sandstrom, K.J. Labori, E.H. Kure, P.M. 184. N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki, T.
Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur, M. Ochiya, Secretory mechanisms and intercellular transfer of
Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, M.S. Brady, O. microRNAs in living cells. J. Biol. Chem. 285, 17442–17452 (2010)
Fodstad, V. Muller, K. Pantel, A.J. Minn, M.J. Bissell, B.A. 185. M. Fabbri, A. Paone, F. Calore, R. Galli, E. Gaudio, R.
Garcia, Y. Kang, V.K. Rajasekhar, C.M. Ghajar, I. Matei, H. Santhanam, F. Lovat, P. Fadda, C. Mao, G.J. Nuovo, N. Zanesi,
Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins de- M. Crawford, G.H. Ozer, D. Wernicke, H. Alder, M.A. Caligiuri,
termine organotropic metastasis. Nature 527, 329–335 (2015) P. Nana-Sinkam, D. Perrotti, C.M. Croce, MicroRNAs bind to
172. S. Keller, J. Ridinger, A.K. Rupp, J.W. Janssen, P. Altevogt, Body toll-like receptors to induce prometastatic inflammatory response.
fluid derived exosomes as a novel template for clinical diagnos- Proc. Natl. Acad. Sci. U. S. A. 109, E2110–E2116 (2012)
tics. J. Transl. Med. 9, 86 (2011) 186. F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard, J.P.
173. C.L. Chen, Y.F. Lai, P. Tang, K.Y. Chien, J.S. Yu, C.H. Tsai, H.W. Remy-Martin, W. Boireau, A. Rouleau, B. Simon, D. Lanneau, A.
Chen, C.C. Wu, T. Chung, C.W. Hsu, C.D. Chen, Y.S. Chang, P.L. De Thonel, G. Multhoff, A. Hamman, F. Martin, B. Chauffert, E.
Chang, Y.T. Chen, Comparative and targeted proteomic analyses Solary, L. Zitvogel, C. Garrido, B. Ryffel, C. Borg, L. Apetoh, C.
of urinary microparticles from bladder cancer and hernia patients. Rebe, F. Ghiringhelli, Membrane-associated Hsp72 from tumor-
J. Proteome Res. 11, 5611–5629 (2012) derived exosomes mediates STAT3-dependent immunosuppres-
174. A. Kannan, R.B. Wells, S. Sivakumar, S. Komatsu, K.P. Singh, B. sive function of mouse and human myeloid-derived suppressor
Samten, J.V. Philley, E.R. Sauter, M. Ikebe, S. Idell, S. Gupta, S. cells. J. Clin. Invest. 120, 457–471 (2010)
250 V. Sundararajan et al.

187. A. Bobrie, S. Krumeich, F. Reyal, C. Recchi, L.F. Moita, M.C. 202. F. Aqil, H. Kausar, A.K. Agrawal, J. Jeyabalan, A.H. Kyakulaga,
Seabra, M. Ostrowski, C. Théry, Rab27a supports exosome- R. Munagala, R. Gupta, Exosomal formulation enhances thera-
dependent and -independent mechanisms that modify the tumor peutic response of celastrol against lung cancer. Exp. Mol.
microenvironment and can promote tumor progression. Cancer Pathol. 101, 12–21 (2016)
Res. 72, 4920–4930 (2012) 203. M.S. Kim, M.J. Haney, Y. Zhao, V. Mahajan, I. Deygen, N.L.
188. M. Ruiz-Martinez, A. Navarro, R.M. Marrades, N. Vinolas, S. Klyachko, E. Inskoe, A. Piroyan, M. Sokolsky, O. Okolie, S.D.
Santasusagna, C. Munoz, J. Ramirez, L. Molins, M. Monzo, Hingtgen, A.V. Kabanov, E.V. Batrakova, Development of
YKT6 expression, exosome release, and survival in non-small cell exosome-encapsulated paclitaxel to overcome MDR in cancer
lung cancer. Oncotarget 7, 51515–51524 (2016) cells. Nanomedicine 12, 655–664 (2016)
189. A. Bosque, L. Dietz, A. Gallego-Lleyda, M. Sanclemente, M. 204. R. Munagala, F. Aqil, J. Jeyabalan, R.C. Gupta, Bovine milk-
Iturralde, J. Naval, M.A. Alava, L. Martinez-Lostao, H.J. derived exosomes for drug delivery. Cancer Lett. 371, 48–61 (2016)
Thierse, A. Anel, Comparative proteomics of exosomes secreted 205. S.C. Jang, O.Y. Kim, C.M. Yoon, D.S. Choi, T.Y. Roh, J. Park, J.
by tumoral Jurkat T cells and normal human T cell blasts unravels Nilsson, J. Lotvall, Y.K. Kim, Y.S. Gho, Bioinspired exosome-
a potential tumorigenic role for valosin-containing protein. mimetic nanovesicles for targeted delivery of chemotherapeutics
Oncotarget 7, 29287–29305 (2016) to malignant tumors. ACS Nano 7, 7698–7710 (2013)
190. C. Federici, F. Petrucci, S. Caimi, A. Cesolini, M. Logozzi, M. 206. K. O'Brien, M.C. Lowry, C. Corcoran, V.G. Martinez, M. Daly, S.
Borghi, S. D'Ilio, L. Lugini, N. Violante, T. Azzarito, C. Majorani, Rani, W.M. Gallagher, M.W. Radomski, R.A. MacLeod, L.
D. Brambilla, S. Fais, Exosome release and low pH belong to a O'Driscoll, miR-134 in extracellular vesicles reduces triple-
framework of resistance of human melanoma cells to cisplatin. negative breast cancer aggression and increases drug sensitivity.
PLoS One 9, e88193 (2014) Oncotarget 6, 32774–32789 (2015)
191. X.Q. Li, J.T. Liu, L.L. Fan, Y. Liu, L. Cheng, F. Wang, H.Q. Yu, J. 207. S. Ohno, M. Takanashi, K. Sudo, S. Ueda, A. Ishikawa, N.
Gao, W. Wei, H. Wang, G.P. Sun, Exosomes derived from Matsuyama, K. Fujita, T. Mizutani, T. Ohgi, T. Ochiya, N.
gefitinib-treated EGFR-mutant lung cancer cells alter cisplatin Gotoh, M. Kuroda, Systemically injected exosomes targeted to
sensitivity via up-regulating autophagy. Oncotarget 7, 24585– EGFR deliver antitumor microRNA to breast cancer cells. Mol.
24595 (2016) Ther. 21, 185–191 (2013)
192. H.G. Zhang, H. Kim, C. Liu, S. Yu, J. Wang, W.E. Grizzle, R.P. 208. K.A. Greco, C.A. Franzen, K.E. Foreman, R.C. Flanigan, P.C.
Kimberly, S. Barnes, Curcumin reverses breast tumor exosomes Kuo, G.N. Gupta, PLK-1 silencing in bladder cancer by sirna
mediated immune suppression of NK cell tumor cytotoxicity. delivered with exosomes. Urology 91, e241–e247 (2016)
Biochim. Biophys. Acta 1773, 1116–1123 (2007) 209. J.L. Munoz, S.A. Bliss, S.J. Greco, S.H. Ramkissoon, K.L. Ligon,
193. T.S. Ramasamy, A.Z. Ayob, H.H. Myint, S. Thiagarajah, F. Amini, P. Rameshwar, Delivery of functional anti-mir-9 by mesenchymal
Targeting colorectal cancer stem cells using curcumin and stem cell-derived exosomes to glioblastoma multiforme cells con-
curcumin analogues: insights into the mechanism of the therapeu- ferred chemosensitivity. Mol. Ther. Nucleic Acids 2, e126 (2013)
tic efficacy. Cancer Cell Int. 15, 96 (2015) 210. H. Qi, C. Liu, L. Long, Y. Ren, S. Zhang, X. Chang, X. Qian, H.
194. S. Amigorena, Cancer immunotherapy using dendritic cell- Jia, J. Zhao, J. Sun, X. Hou, X. Yuan, C. Kang, Blood exosomes
derived exosomes. Medicina (B Aires) 60 Suppl 2, 51–54 (2000) endowed with magnetic and targeting properties for cancer thera-
195. G.G. Romagnoli, B.B. Zelante, P.A. Toniolo, I.K. Migliori, J.A.M. py. ACS Nano 10, 3323–3333 (2016)
Barbuto, Dendritic cell-derived exosomes may be a tool for cancer 211. Y. Tian, S. Li, J. Song, T. Ji, M. Zhu, G.J. Anderson, J. Wei, G.
immunotherapy by converting tumor cells into immunogenic tar- Nie, A doxorubicin delivery platform using engineered natural
gets. Front. Immunol. 5, 692 (2014) membrane vesicle exosomes for targeted tumor therapy.
196. Q. Rao, B. Zuo, Z. Lu, X. Gao, A. You, C. Wu, Z. Du, H. Yin, Biomaterials 35, 2383–2390 (2014)
Tumor-derived exosomes elicit tumor suppression in murine he- 212. T. Yang, P. Martin, B. Fogarty, A. Brown, K. Schurman, R.
patocellular carcinoma models and human in vitro. Hepatology Phipps, V.P. Yin, P. Lockman, S. Bai, Exosome delivered antican-
64, 456–472 (2016) cer drugs across the blood-brain barrier for brain cancer therapy in
197. J. Wang, L. Wang, Z. Lin, L. Tao, M. Chen, More efficient induc- Danio rerio. Pharm. Res. 32, 2003–2014 (2015)
tion of antitumor T cell immunity by exosomes from CD40L gene- 213. C.S. Hong, L. Muller, M. Boyiadzis, T.L. Whiteside, Isolation and
modified lung tumor cells. Mol. Med. Rep. 9, 125–131 (2014) characterization of CD34+ blast-derived exosomes in acute mye-
198. M. Damo, D.S. Wilson, E. Simeoni, J.A. Hubbell, TLR-3 stimu- loid leukemia. PLoS One 9, e103310 (2014)
lation improves anti-tumor immunity elicited by dendritic cell 214. C.J. Beckham, J. Olsen, P.N. Yin, C.H. Wu, H.J. Ting, F.K. Hagen,
exosome-based vaccines in a murine model of melanoma. Sci. E. Scosyrev, E.M. Messing, Y.F. Lee, Bladder cancer exosomes
Rep. 5, 17622 (2015) contain EDIL-3/Del1 and facilitate cancer progression. J. Urol.
199. Y. Xie, O. Bai, H. Zhang, J. Yuan, S. Zong, R. Chibbar, K. Slattery, 192, 583–592 (2014)
M. Qureshi, Y. Wei, Y. Deng, J. Xiang, Membrane-bound HSP70- 215. C. Berrondo, J. Flax, V. Kucherov, A. Siebert, T. Osinski, A.
engineered myeloma cell-derived exosomes stimulate more effi- Rosenberg, C. Fucile, S. Richheimer, C.J. Beckham, Expression
cient CD8(+) CTL- and NK-mediated antitumour immunity than of the long non-coding rna hotair correlates with disease progres-
exosomes released from heat-shocked tumour cells expressing cy- sion in bladder cancer and is contained in bladder cancer patient
toplasmic HSP70. J. Cell. Mol. Med. 14, 2655–2666 (2010) urinary exosomes. PLoS One 11, e0147236 (2016)
200. L.H. Lv, Y.L. Wan, Y. Lin, W. Zhang, M. Yang, G.L. Li, H.M. Lin, 216. C. Eichelser, I. Stuckrath, V. Muller, K. Milde-Langosch, H.
C.Z. Shang, Y.J. Chen, J. Min, Anticancer drugs cause release of Wikman, K. Pantel, H. Schwarzenbach, Increased serum levels
exosomes with heat shock proteins from human hepatocellular of circulating exosomal microRNA-373 in receptor-negative
carcinoma cells that elicit effective natural killer cell antitumor breast cancer patients. Oncotarget 5, 9650–9663 (2014)
responses in vitro. J. Biol. Chem. 287, 15874–15885 (2012) 217. S. Khan, H.F. Bennit, D. Turay, M. Perez, S. Mirshahidi, Y. Yuan,
201. G. Fuhrmann, A. Serio, M. Mazo, R. Nair, M.M. Stevens, Active N.R. Wall, Early diagnostic value of survivin and its alternative
loading into extracellular vesicles significantly improves the cel- splice variants in breast cancer. BMC Cancer 14, 176 (2014)
lular uptake and photodynamic effect of porphyrins. J. Control. 218. I. Vardaki, S. Ceder, D. Rutishauser, G. Baltatzis, T. Foukakis,
Release 205, 35–44 (2015) T. Panaretakis, Periostin is identified as a putative metastatic
The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications 251

marker in breast cancer-derived exosomes. Oncotarget 7, mutated KRAS and p53 DNA in the serum exosomes of patients
74966–74978 (2016) with pancreatic cancer. J. Biol. Chem. 289, 3869–3875 (2014)
219. J. Liu, H. Sun, X. Wang, Q. Yu, S. Li, X. Yu, W. Gong, Increased 234. S.A. Melo, L.B. Luecke, C. Kahlert, A.F. Fernandez, S.T.
exosomal microRNA-21 and microRNA-146a levels in the Gammon, J. Kaye, V.S. LeBleu, E.A. Mittendorf, J. Weitz, N.
cervicovaginal lavage specimens of patients with cervical cancer. Rahbari, C. Reissfelder, C. Pilarsky, M.F. Fraga, D. Piwnica-
Int. J. Mol. Sci. 15, 758–773 (2014) Worms, R. Kalluri, Glypican-1 identifies cancer exosomes and
220. T. Matsumura, K. Sugimachi, H. Iinuma, Y. Takahashi, J. detects early pancreatic cancer. Nature 523, 177–182 (2015)
Kurashige, G. Sawada, M. Ueda, R. Uchi, H. Ueo, Y. Takano, 235. P.J. Mitchell, J. Welton, J. Staffurth, J. Court, M.D. Mason, Z.
Y. Shinden, H. Eguchi, H. Yamamoto, Y. Doki, M. Mori, T. Tabi, A. Clayton, Can urinary exosomes act as treatment response
Ochiya, K. Mimori, Exosomal microRNA in serum is a novel markers in prostate cancer? J. Transl. Med. 7, 4 (2009)
biomarker of recurrence in human colorectal cancer. Br. J. 236. T. Kato, K. Mizutani, K. Kameyama, K. Kawakami, Y. Fujita, K.
Cancer 113, 275–281 (2015) Nakane, Y. Kanimoto, H. Ehara, H. Ito, M. Seishima, T. Deguchi,
221. H. Ogata-Kawata, M. Izumiya, D. Kurioka, Y. Honma, Y. M. Ito, Serum exosomal P-glycoprotein is a potential marker to
Yamada, K. Furuta, T. Gunji, H. Ohta, H. Okamoto, H. Sonoda, diagnose docetaxel resistance and select a taxoid for patients with
M. Watanabe, H. Nakagama, J. Yokota, T. Kohno, N. Tsuchiya, prostate cancer. Urol. Oncol. 33, e315–e320 (2015)
Circulating exosomal microRNAs as biomarkers of colon cancer. 237. X. Huang, T. Yuan, M. Liang, M. Du, S. Xia, R. Dittmar, D. Wang,
PLoS One 9, e92921 (2014) W. See, B.A. Costello, F. Quevedo, W. Tan, D. Nandy, G.H.
222. Q. Li, Y. Shao, X. Zhang, T. Zheng, M. Miao, L. Qin, B. Wang, G. Bevan, S. Longenbach, Z. Sun, Y. Lu, T. Wang, S.N. Thibodeau,
Ye, B. Xiao, J. Guo, Plasma long noncoding RNA protected by L. Boardman, M. Kohli, L. Wang, Exosomal miR-1290 and miR-
exosomes as a potential stable biomarker for gastric cancer. 375 as prognostic markers in castration-resistant prostate cancer.
Tumour Biol. 36, 2007–2012 (2015) Eur. Urol. 67, 33–41 (2015)
223. H. Wang, L. Hou, A. Li, Y. Duan, H. Gao, X. Song, Expression of 238. James Graham Brown Cancer Center. Phase I clinical trial inves-
serum exosomal microRNA-21 in human hepatocellular carcino- tigating the ability of plant exosomes to deliver curcumin to nor-
ma. Biomed. Res. Int. 2014, 864894 (2014) mal and malignant colon tissue. http://clinicaltrials.gov/show/
224. K. Sugimachi, T. Matsumura, H. Hirata, R. Uchi, M. Ueda, H. NCT01294072. Accessed 16 September 2016
Ueo, Y. Shinden, T. Iguchi, H. Eguchi, K. Shirabe, T. Ochiya, Y. 239. James Graham Brown Cancer Center. preliminary clinical trial
Maehara, K. Mimori, Identification of a bona fide microRNA investigating the ability of plant exosomes to abrogate oral muco-
biomarker in serum exosomes that predicts hepatocellular carci- sitis induced by combined chemotherapy and radiation in head
noma recurrence after liver transplantation. Br. J. Cancer 112, and neck cancer patients. http://clinicaltrials.gov/show/
532–538 (2015) NCT01668849. Accessed 16 September 2016
225. J. Wang, Y. Zhou, J. Lu, Y. Sun, H. Xiao, M. Liu, L. Tian, 240. B. Besse, M. Charrier, V. Lapierre, E. Dansin, O. Lantz, D.
Combined detection of serum exosomal miR-21 and HOTAIR Planchard, T. Le Chevalier, A. Livartoski, F. Barlesi, A.
as diagnostic and prognostic biomarkers for laryngeal squamous Laplanche, S. Ploix, N. Vimond, I. Peguillet, C. Thery, L.
cell carcinoma. Med. Oncol. 31, 148 (2014) Lacroix, I. Zoernig, K. Dhodapkar, M. Dhodapkar, S. Viaud,
226. M. Guan, X. Chen, Y. Ma, L. Tang, L. Guan, X. Ren, B. Yu, W. J.C. Soria, K.S. Reiners, E. Pogge von Strandmann, F. Vely, S.
Zhang, B. Su, MDA-9 and GRP78 as potential diagnostic bio- Rusakiewicz, A. Eggermont, J.M. Pitt, L. Zitvogel, N. Chaput,
markers for early detection of melanoma metastasis. Tumour Dendritic cell-derived exosomes as maintenance immunotherapy
Biol. 36, 2973–2982 (2015) after first line chemotherapy in NSCLC. Oncoimmunology 5,
227. E. Alegre, M.F. Sanmamed, C. Rodriguez, O. Carranza, S. Martin- e1071008 (2016)
Algarra, A. Gonzalez, Study of circulating microRNA-125b levels 241. S. Viaud, M. Terme, C. Flament, J. Taieb, F. Andre, S. Novault, B.
in serum exosomes in advanced melanoma. Arch. Pathol. Lab. Escudier, C. Robert, S. Caillat-Zucman, T. Tursz, L. Zitvogel, N.
Med. 138, 828–832 (2014) Chaput, Dendritic cell-derived exosomes promote natural killer
228. J. Klibi, T. Niki, A. Riedel, C. Pioche-Durieu, S. Souquere, E. cell activation and proliferation: a role for NKG2D ligands and
Rubinstein, S. Le Moulec, J. Guigay, M. Hirashima, F. Guemira, IL-15Ralpha. PLoS One 4, e4942 (2009)
D. Adhikary, J. Mautner, P. Busson, Blood diffusion and Th1- 242. B. Escudier, T. Dorval, N. Chaput, F. Andre, M.P. Caby, S.
suppressive effects of galectin-9-containing exosomes released Novault, C. Flament, C. Leboulaire, C. Borg, S. Amigorena, C.
by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Boccaccio, C. Bonnerot, O. Dhellin, M. Movassagh, S. Piperno,
Blood 113, 1957–1966 (2009) C. Robert, V. Serra, N. Valente, J.B. Le Pecq, A. Spatz, O. Lantz,
229. Y. Li, Y. Zhang, F. Qiu, Z. Qiu, Proteomic identification of T. Tursz, E. Angevin, L. Zitvogel, Vaccination of metastatic mel-
exosomal LRG1: a potential urinary biomarker for detecting anoma patients with autologous dendritic cell (DC) derived-
NSCLC. Electrophoresis 32, 1976–1983 (2011) exosomes: results of thefirst phase I clinical trial. J. Transl. Med.
230. Y. Tanaka, H. Kamohara, K. Kinoshita, J. Kurashige, T. Ishimoto, 3, 10 (2005)
M. Iwatsuki, M. Watanabe, H. Baba, Clinical impact of serum 243. Exosome Diagnostics, Inc. Clinical validation of a urinary
exosomal microRNA-21 as a clinical biomarker in human esoph- exosome gene signature in men presenting for suspicion of pros-
ageal squamous cell carcinoma. Cancer 119, 1159–1167 (2013) tate cancer. http://www.clinicaltrials.gov/show/NCT02702856.
231. D.D. Taylor and C. Gercel-Taylor, MicroRNA signatures of Accessed 16 September 2016
tumor-derived exosomes as diagnostic biomarkers of ovarian can- 244. Hospital Miguel Servet. Circulating exosomes as potential prog-
cer. Gynecol. Oncol. 110, 13–21 (2008) nostic and predictive biomarkers in advanced gastric cancer pa-
232. X. Ying, Q. Wu, X. Wu, Q. Zhu, X. Wang, L. Jiang, X. Chen and tients. http://www.clinicaltrials.gov/show/NCT01779583.
X. Wang, Epithelial ovarian cancer-secreted exosomal miR-222- Accessed 16 September 2016
3p induces polarization of tumor-associated macrophages. 245. New Mexico Cancer Care Alliance. An observational, single-
Oncotarget 7, 43076–43087 (2016) institution pilot/feasibility study of exosome testing as a screening
233. C. Kahlert, S.A. Melo, A. Protopopov, J. Tang, S. Seth, M. Koch, modality for human papillomavirus-positive oropharyngeal squa-
J. Zhang, J. Weitz, L. Chin, A. Futreal, R. Kalluri, Identification of mous cell carcinoma. http://clinicaltrials.gov/show/
double-stranded genomic DNA spanning all chromosomes with NCT02147418. Accessed 16 September 2016
252 V. Sundararajan et al.

246. Centre Georges Francois Leclerc. Pilot study with the aim to quan- 251. Centre Hospitalier Universitaire de Nice. Pilot study of exosomes
tify a stress protein in the blood and in the urine for early diagnosis before and after braf inhibitor therapy in patients with advanced
of malgnant solid tumors. http://clinicaltrials.gov/show/ unresectable or metastatic braf mutation-positive melanoma.
NCT02662621. Accessed 16 September 2016 http://www.clinicaltrials.gov/show/NCT02310451. Accessed 16
247. National Taiwan University Hospital. Anaplastic thyroid cancer September 2016
and follicular thyroid cancer-derived exosomal analysis via treat- 252. Midwest Biomedical Research Foundation. Evaluation of
ment of lovastatin and vildagliptin and pilot prognostic study via microrna expression in blood and cytology specimens as a
urine exosomal biological markers in thyroid cancer patients. novel method for detecting barrett's esophagus. http://www.
http://clinicaltrials.gov/show/NCT02862470. Accessed 16 clinicaltrials.gov/show/NCT02464930. Accessed 16
September 2016 September 2016
248. Thomas Jefferson University. Phase 1 study in humans evaluating 253. Xinqiao Hospital of Chongqing. Clinical research for the consis-
the safety of rectus sheath implantation of diffusion chambers tency analysis of pd-l1 in cancer tissue and plasma exosome.
encapsulating autologous malignant glioma cells treated with http://www.clinicaltrials.gov/show/NCT02890849. Accessed 16
insulin-like growth factor receptor-1 antisense September 2016
oligodeoxynucleotide in 12 patients with recurrent malignant gli- 254. Xinqiao Hospital of Chongqing. Clinical research for the consis-
oma. http://www.clinicaltrials.gov/show/NCT01550523. tency analysis of pd-l1 in lung cancer tissue and plasma exosome
Accessed 16 September 2016 before and after radiotherapy. http://www.clinicaltrials.gov/show/
249. Thomas Jefferson University. Phase I study in humans evaluating NCT02869685. Accessed 16 September 2016
the safety of rectus sheath implantation of diffusion chambers en- 255. Memorial Sloan Kettering Cancer Center. Interrogation of
capsulating autologous malignant glioma cells treated with insulin- exosome-mediated intercellular signaling in patients with pancre-
like growth factor receptor-1 antisense oligodeoxynucleotide (igf-1r/ atic cancer. http://www.clinicaltrials.gov/show/NCT02393703.
as odn) in 32 patients with newly diagnosed malignant glioma. Accessed 16 September 2016
http://clinicaltrials.gov/show/NCT02507583. Accessed 16
256. Centre Oscar Lambret. Early biomarkers of tumor response in
September 2016
high dose hypofractionated radiotherapy word package 3: immune
250. S. Dai, D. Wei, Z. Wu, X. Zhou, X. Wei, H. Huang, G. Li, I.
response. http://clinicaltrials.gov/show/NCT02439008. Accessed
Phase, clinical trial of autologous ascites-derived exosomes
16 September 2016
combined with GM-CSF for colorectal cancer. Mol. Ther. 16,
782–790 (2008)

You might also like