Exosomes 4

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

G Model

YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS


Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cell & Developmental Biology


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

Review

Exosomes: Key mediators of metastasis and pre-metastatic niche


formation
Richard J. Lobb a,1 , Luize G. Lima a,1 , Andreas Möller a,b,∗
a
Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
b
School of Medicine, University of Queensland, Brisbane, QLD 4072, Australia

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

Article history: While tumour cells are classically known to communicate via direct cell-to-cell contact and the secretion
Received 16 September 2016 of soluble protein-based factors such as cytokines and growth factors, alternative novel mechanisms that
Received in revised form promote tumour progression have recently emerged. Now, new critical components of the secretome
22 December 2016
thought to be involved in tumour progression are exosomes, small vesicles of endocytic origin that carry
Accepted 6 January 2017
a variety of bioactive molecules, including proteins, lipids, RNA, as well as DNA molecules. Cancer cell-
Available online xxx
derived exosomes have been shown to participate in crucial steps of metastatic spread of a primary
tumour, ranging from oncogenic reprogramming of malignant cells to formation of pre-metastatic niches.
Keywords:
Cancer
These effects are achieved through the mediation of intercellular cross-talk and subsequent modification
Extracellular vesicles of both local and distant microenvironments in an autocrine and paracrine fashion. Here, we summarise
Pre-metastatic niche the recent findings that implicate this non-canonical signalling within the tumour as a critical driver
Metastasis of metastatic disease progression, and discuss how understanding the molecular mechanisms involved
Exosomes in exosome-mediated metastasis is of great value for the development of new therapeutic strategies to
prevent cancer progression.
© 2017 Published by Elsevier Ltd.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. The role of exosomes in the primary tumour microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Exosomes and angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Oncogenic transfer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3. Exosomes and immune suppression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Exosome-related pro-metastatic mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1. The role of tumour-exosomes in pre-metastatic niche formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.2. Exosomes and vascular remodelling in pre-metastatic niches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.3. Effects of cancer-derived exosomes on other non-immune cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.4. Impact of cancer-derived exosomes on immune cells in pre-metastatic niches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.5. Modification of the metabolic environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.6. Determination of organotropic homing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .00
3.7. Non-tumour-derived exosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Abbreviation: BM, bone marrow; BMDCs, bone marrow-derived cells; EVs, extracellular vesicles; FasL, fas ligand; FGF2, fibroblast growth factor 2; GM-CSF, granulocyte
macrophage colony-stimulating factor; HGF, hepatocyte growth factor; HIF-1, hypoxia-inducible factor 1; HUVECs, human umbilical vein endothelial cells; IBA-1, ionized
calcium-binding adapter molecule 1; IL-6, interleukin 6; MDSCs, myeloid-derived suppressor cells; MMP, matrix metalloproteinase; MVBs, multivesicular bodies; PGE2,
prostaglandin E2; PKM, pyruvate kinase; PlGF, placental growth factor; PTEN, Phosphatase and tensin homolog; TGF-␤, transforming growth factor beta; TLR, toll like
receptor; TNF-␣, tumour necrosis factor alpha; TRAIL, TNF-related apoptosis-inducing ligand; VEGF, vascular endothelial growth factor; VEGFR-1, vascular endothelial
growth factor receptor 1; VLA-4, very late antigen 4; ZO-1, zonula occludens.
∗ Corresponding author at: 300 Herston Road, Herston, QLD, 4006, Australia.
E-mail address: andreas.moller@qimrberghofer.edu.au (A. Möller).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.semcdb.2017.01.004
1084-9521/© 2017 Published by Elsevier Ltd.

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
2 R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

4. Clinical utility of exosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction systemically modify distant organs to provide favourable microen-


vironments for the dissemination and outgrowth of metastases
Despite significant advancements in therapies, a large propor- [13–15]. These effects are achieved by the bioactive molecular
tion of cancer patients will ultimately pass away due to metastatic constituents of exosomes mentioned earlier, which are in turn
disease progression. It is therefore a key goal of cancer research determined by the cell of origin that the exosomes are secreted
efforts to further our understanding of metastatic disease progres- from [10,13–15]. For this very reason, exosomes have become a
sion and its dynamics. Based on recent research findings, it is clear valuable target in identifying novel cancer biomarkers that could
that metastatic disease is not a random process. That is, the seeding potentially diagnose cancer, predict patient outcome or response,
of cancer cells at secondary organs does occur in an organ-specific and understand, and perhaps prevent, cancer progression.
manner, depending on the cancer type, verifying the so called “seed Cancer cell-derived exosomes have been shown to significantly
and soil” hypothesis, which was originally proposed over 100 years contribute to different aspects of metastatic dissemination of a
ago by Stephen Paget [1,2]. This concept of metastatic growth speci- primary tumour, including invasion of the surrounding tissues,
ficity has since been validated experimentally and clinically [3], angiogenesis, modulation of immune responses, and formation of
and decisively shown that even when cancer cells can be found the pre-metastatic niche [16]. The roles exosomes have in pre-
in the vasculature of multiple organs, only selective sites consis- metastatic niche formation are diverse, and range from metabolic
tently develop metastatic tumour deposits [4]. It is now widely reprogramming, to recruitment of numerous immune and non-
accepted that the spread of cancer cells to secondary organs is immune stromal cells in order to facilitate metastatic outgrowth.
indeed promoted by the prior formation of a specialised environ- The aim of this review is to discuss the particular multi-faceted roles
ment at distant sites, termed the pre-metastatic niche. of these extracellular particles that impact both cancer cell spread
Originally, pre-metastatic niche formation was shown to consist from primary to secondary organs, resulting in the outgrowth of
of bone marrow-derived hematopoietic progenitor cells (BMDCs), macroscopic metastatic lesions (Fig. 1), and lastly, provide new
which accumulate at pre-metastatic sites in organs different to the insights into how this can be translated to clinical applications.
site of the primary tumour and before the arrival of any cancer cells
[5]. These BMDCs expressed VLA-4, and were recruited by tumour- 2. The role of exosomes in the primary tumour
derived secreted factors, which resulted in the accumulation of the microenvironment
VLA-4 ligand fibronectin at pre-metastatic organs [5]. Since then,
numerous studies have established that variable tumour-secreted 2.1. Exosomes and angiogenesis
factors are responsible for establishing a permissive secondary
organ through the modulation of a number of stromal cell-types [6]. A common process known to be involved in tumour progres-
However, these studies have largely focused on canonical intercel- sion is hypoxia. Hypoxia is the relative reduction in oxygen tension
lular communication based on classical cytokines, chemokines and and occurs in all solid tumours larger than 1 cm3 . This results
growth factors; yet, it is becoming evident non-canonical signalling from inadequate blood supply due to the aberrant microcirculation
pathways are also involved in pre-metastatic niche formation [7]. found in most tumours [17,18]. The microcirculation is essential
More recently, a new mechanism of intercellular communi- in normal tissues homeostasis, as it is responsible for balancing
cation via the secretion of extracellular vesicles (EVs) has been the supply of nutrients and removal of cellular waste products
established as an integral part of complex tumour-host interac- [19,20]. In the context of a solid tumour, neo-angiogenesis, or
tions [8]. Clinical interest in EVs has heightened due to the fact that the physiological process of new blood vessel growth, is neces-
continuing evidence supports the notion that EVs represent a new sary for continued tumour growth in response to hypoxia [19,20].
paradigm of intercellular communication [9]. EVs are constitutively Typically, solid tumours have been described to induce vascu-
secreted by most likely all different cell lineages in the body, and lar remodelling through the secretion of soluble factors, such as
carry diverse molecular contents, including proteins, lipids, RNA VEGF and PIGF. However, exosomes are also capable of being
(mRNA, miRNA, lncRNA, and other RNA molecules), as well as DNA key mediators between cancer cells and the surrounding vas-
molecules (dsDNA, ssDNA, mtDNA) [10]. There are a variety of EVs culature by eliciting pro-angiogenic responses [21,22]. Hypoxic
subtypes, and their accurate annotation is currently an ongoing, glioblastoma-derived exosomes are capable of inducing microvas-
contentious problem in the field [11]. There are clearly two dis- culature sprouting and vascularisation of xenografts models [22].
tinct subtypes of EVs that can be separated effectively through In response to this angiogenic phenotype, tumours displayed accel-
differential centrifugation: microvesicles are the larger subclass erated growth kinetics, demonstrating a role for exosomes during
(200–1000 nm) and sediment between 10,000 and 20,000g, while early tumour progression events. Hypoxia content changes in
smaller EVs known as exosomes exhibit a more homogenous size multiple myeloma derived exosomes also induce angiogenesis in
distribution of 30–150 nm, and sediment at 100,000g [9,11,12]. recipient endothelial cells [23]. Under hypoxia conditions, multiple
Exosomes are constitutively released by a variety of cell lineages, myeloma cells increase the secretion of miR-135b contained within
including cancer cells [10]. The biogenesis of exosomes is classically exosomes in order to promote an angiogenic response. Endothelial
described as originating from the endosomal compartment and are cells that received miR-135b-containing exosomes had signifi-
formed as intraluminal vesicles by inward budding of the limit- cantly increased HIF-1 alpha levels, as miR-135b binds the 3 -UTR of
ing membrane of late endosomes, forming multivescular bodies factor-inhibiting hypoxia-inducible factor-1 (FIH-1). This induced
(MVBs). The subsequent fusion of MVBs with the plasma membrane a hypoxic response and greatly accelerated angiogenesis [23]. In
allows the release of exosomes into the extracellular environment. further support of the role that exosomes play during angiogene-
This fusion and release into the extracellular milieu, allows can- sis, CD105+ vesicles from renal carcinoma cells have been shown
cer cells to utilise exosomes to modify local microenvironments, or to induce proliferation of HUVECs in vitro and in vivo [21]. These

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 3

Fig. 1. Exosomal mechanisms of metastasis promotion. (a) Exosomes secreted by cancer cells contribute to the intercellular cross-talk and subsequent reprogramming of
the primary tumour microenvironment in an autocrine and paracrine fashion. Bioactive content of exosomes have been implicated in different aspects of primary tumour
progression, including angiogenesis, modulation of immune responses, and oncogenic reprogramming. (b) Moreover, exosomal factors are capable of mobilising bone
marrow-derived cells (BMDCs) to the primary tumour, which in turn facilitate the establishment of a supportive microenvironment. (c) This non-canonical signalling within
the primary tumour has also emerged as a critical driver of pre-metastatic niche formation. Tumour-derived exosomes can home to specific secondary organs and directly
modify the local microenvironment by interacting with distinct resident cell populations. A variety of factors contained within exosomes were shown to participate in a range
of biological processes, from modulation of immune and non-immune stromal cells to metabolic reprogramming. (d) Chemokines and other pro-inflammatory molecules
are also upregulated in pre-metastatic sites in response to exosome-mediated signalling, leading to the recruitment and activation of BMDCs. Altogether, exosome-induced
pre-metastatic niche modifications generate a microenvironment conducive to metastatic outgrowth (e), thereby facilitating cancer cell dissemination and colonisation of
distant tissues in an organotropic specific manner.

data provide novel insights into mechanisms of how tumours can association between inflammation and tumourigenesis [29,30].
circumvent the clinical inhibition of angiogenic proteins (for exam- However, inflammation can have paradoxical roles during tumour
ple through VEGF-neutralizing antibodies), and induce vascular progression, the outcome depending on the balance between
remodelling, allowing tumour-induced neo-angiogenesis through tumour-promoting and tumour-suppressing activities in the local
exosomal transfer. microenvironment [30]. Classically, chemokines and cytokines
secreted through canonical pathways have been thought of as
the master regulators of inflammatory processes in the tumour
2.2. Oncogenic transfer
microenvironment, yet it has recently emerged that exosomes add
another layer of complexity to cancer-associated inflammatory
Tumours typically consist of numerous subclones that are genet-
reactions. Cancer cell-derived exosomes contain molecular con-
ically and phenotypically different from one another [24]. It is
stituents that can induce immune cell dysfunction and contribute
therefore possible that different subclones can transfer onco-
to silencing of anti-tumour immune responses by several different
genic traits via exosomes to other cancer cells in the primary
ways [31], either by directly altering immune cell function, or indi-
tumour microenvironment. Indeed, glioma cells have been shown
rectly through modifying non-immune cell stromal phenotypes.
to enhance tumourigenesis of recipient cells through the transfer
Tumour-derived exosomes can suppress T cell immunity and
of mutant epidermal growth factor receptor (EGFRvIII) [25]. Recip-
direct immune cells towards a tumour-promoting phenotype. The
ient cells were then shown to have a higher proliferation index and
effects on T cells are partly achieved through causing apopto-
were more resistant to apoptosis. Mutant KRAS can also be trans-
sis of cytotoxic T-cells via the transfer of the death ligands FasL
ferred via exosomes and increases the three-dimensional growth
and TRAIL [32,33]. Furthermore, exosomes are capable of editing
of wild-type recipient colon cells [26]. Furthermore, metastatic
the immune composition of the primary tumour microenviron-
melanoma-derived exosomes are capable of inducing a metastatic
ment by recruiting and expanding myeloid-derived suppressor
phenotype of non-metastatic melanoma recipient cells [27]. It
cells (MDSCs). These cells have been identified in both humans and
remains unclear whether this protein inheritance changes tumour
mice as a heterogeneous population of immature myeloid cells,
growth and metastasis in a permanent or an acute manner, yet it
capable of mediating immunosuppressive functions within the
provides interesting insights into how tumour cells can communi-
tumour microenvironment [34]. In mice, MDSC subsets are char-
cate and promote tumour progression through exosomal transfer
acterised by the expression of CD11b and Gr-1, while in humans
of oncogenic proteins.
they are usually identified by a CD11b+ /CD33+ /HLA-DR- phenotype
[34]. Breast carcinoma-derived exosomes containing prostaglandin
2.3. Exosomes and immune suppression E2 and TGF-␤ skew the development of myeloid cells toward
a MDSC phenotype, thus promoting tumour development [35].
Rudolf Virchow was the first to observe leukocyte infiltration Whether these and other cytokines/chemokines are indeed con-
into tumours in the 19th century [28]. Since then, experi- tained in tumour-derived exosomes or are merely contaminants
mental and clinical investigations have demonstrated the close

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
4 R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

from either tumour or non-tumour cell sources, caused by the exo- [42]. The term ‘pre-metastatic niche’ was coined later by demon-
some isolation procedure, is however still a matter of debate [36]. strating that tumour-derived conditioned media alone is capable
Nonetheless, these data collectively suggest that exosomes play of inducing migration of VEGFR-1-expressing BMDCs into organ-
a key role in deciding the tumour-promoting fate of infiltrating specific pre-metastatic sites, as well as posterior formation of
immune cells. BMDCs clusters that precede tumour cell arrival [5]. These clusters,
Tumour-derived exosomes can further promote an immune- in turn, create a favourable microenvironment for incoming tumour
suppressed tumour microenvironment through the expansion of cells, in a mechanism dependent on the expression of fibronectin,
CD4+ /CD25+ regulatory T cells [37]. This is mechanistically linked VLA-4, Id3, and MMP-9 [5]. Cell-free conditioned medium from
to the functional expression of CD39 and CD73 on exosomes, and hypoxic breast cancers was also shown to drive the accumulation of
consequent ATP-dependent adenosine production in recipient cells a specific subset of BMDCs, known as CD11b+ MDSCs, in the lungs of
[38]. Melanoma exosomes also impair dendritic cell (DC) differ- immune-competent mice [43]. There, the formation of an immune
entiation from monocytes, and simultaneously convert these cells suppressed environment through the reduced cytotoxic capacity
toward an immune-suppressive phenotype [39]. In support of these of Natural Killer (NK) cells, in turn promote the metastatic out-
observations, an increased frequency of CD14+ /HLA-DR− myeloid growth of both breast cancer and melanoma tumours [43]. Since
cells associated with a TGF-␤-mediated inhibitory activity on T then, a variety of BMDC lineages, among other organ resident cell
cells was found in the peripheral blood of melanoma patients [39]. lineages, have been shown to contribute to the generation of pre-
Even though these studies have focused on the role of immune metastatic niches, and a large number of chemokines, cytokines
cell uptake in the primary tumour microenvironment, it is inter- and growth factors have been connected to specific pre-metastatic
esting to speculate that similar mechanisms could be happening at niche-related processes [44,45]. In recent times, exosomes have
pre-metastatic organs. been described to have unique and central functions during pre-
Finally, it seems that modulation of immune cells behaviour by metastatic niche establishment and maintenance.
cancer-derived exosomes can also be triggered by other mecha-
nisms, such as transfer of functional mRNA between tumour and 3.2. Exosomes and vascular remodelling in pre-metastatic niches
host cells [40]. Transplanting glioma and lung carcinoma cells,
engineered to express Cre recombinase, into mice with a Cre- Modulation of vascular permeability, and stimulation of
lox background, the uptake of Cre mRNA-contaning exosomes by neo-angiogenesis, are key steps during pre-metastatic niche estab-
CD11b+ Gr-1+ MDSCs present in the primary tumour microenvi- lishment that favour the initial extravasation and subsequent
ronment could be tracked [40]. It is noteworthy that recombined metastatic growth of tumour cells into secondary organs [45]. The
MDSCs showed alterations in both their immunosuppressive phe- ability to modulate endothelial cell activity could also represent a
notype and miRNA profile [40]. These observations suggest a crucial element in the repertoire of pro-metastatic roles of tumour-
long-term, stable genetic reprogramming of myeloid cells by exo- derived exosomes. Recently, the importance of extracellular miRNA
somes. contained within exosomes in vascular remodelling has been
highlighted. Metastatic MDA-MB-231 breast cancer cells secrete
exosomes enriched for the miRNA miR-105 [46]. The presence of
3. Exosome-related pro-metastatic mechanisms
this miRNA in breast cancer-derived exosomes increases metastasis
by facilitating endothelial cell barrier destruction through down-
In earlier studies, the majority of cancer research has focused
regulation of the tight junction zonula occludens 1 (ZO-1) protein.
on the pathogenesis of the primary tumour. However, metastatic
The reduction of ZO-1 by miR-105 increases endothelial barrier per-
disease is responsible for over 90% of cancer-related deaths, and
meability and facilitates cancer cell migration in vitro and in vivo.
very few therapies have proven successful in the clinic for treat-
Perturbation of vascular cell barrier function is a key step in allow-
ing patients with metastatic deposits [41]. Therefore, we need
ing cancer cells to not only intravasate into the circulation, but also
to understand the underlying mechanisms of metastasis in order
extravasate at secondary sites to form metastatic lesions. Indeed,
to generate novel, effective therapies for patients with advanced,
exosomal miR-105 increased vascular permeability at secondary
aggressive cancers. The metastatic process is composed of a number
organs, thereby increasing metastasis by facilitating colonisation
of sequential events that tumour cells are required to accomplish
of new tissues [46].
in order to successfully disseminate to secondary organs. Known
In support of these observations, exosomes purified from the
as the metastatic cascade, a series of steps involving local inva-
supernatant of CD105+ renal cancer stem cells that induced vascu-
sion, survival and evasion of immune responses, intravasation into
lar remodelling by increasing angiogenesis at the primary tumour,
the circulation, and extravasation at secondary organs are required
also induced an angiogenic response at pre-metastatic sites [21].
for the formation of macroscopic metastatic lesions. Importantly,
CD105+ vesicles enhance lung metastasis in vivo due to the
exosomes are capable of enhancing the systematic entry and
uptake and parallel up-regulation of VEGF and MMP-2 in lung
progression of cancer cells along the metastatic cascade [19,20],
endothelial cells [21], thereby demonstrating the importance of
highlighting the importance of understanding exosome biology
vascular remodelling at pre-metastatic organs. Melanoma-derived
in metastatic disease progression. In the following paragraphs,
exosomes also accumulated at common sites of melanoma metas-
we will further examine the role of cancer-derived exosomes in
tasis, and subsequently caused vascular remodelling by enhancing
diverse mechanisms relating to metastasis, which can orchestrate
endothelial cell permeability and up-regulation of the expression of
pre-metastatic niche establishment at secondary organs and con-
many genes previously associated with a microenvironment con-
sequent metastatic outgrowth (Fig. 1).
ducive to metastatic outgrowth [15].
Taken together, these results demonstrate that tumour exo-
3.1. The role of tumour-exosomes in pre-metastatic niche somes exhibit a strong impact on endothelial cell function, which
formation could contribute to remodelling of the microenvironment in order
to promote tumour progression. It is noteworthy, however, that
Changes at secondary organs prior to tumour cell metastasis induction of vascular permeability by tumour exosomes in dis-
were first described to be caused by matrix metalloproteinase tant organs, such as the lung, is not always accompanied by lung
9 (MMP-9), which is specifically induced in pre-metastatic lung parenchyma uptake or facilitation of metastasis [14]. These obser-
endothelial cells and macrophages by distant primary tumour cells vations indicate that vascular remodelling alone is not sufficient

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 5

to promote tumour outgrowth in secondary organs, although it is Exosomes derived from breast cancer cells were shown to be
regarded as a necessary step during the metastatic cascade. preferentially taken up by CD11b+ Gr-1+ BMDCs, which in turn
accumulated in the spleen and lung of exosome-treated mice [57].
Further in vitro assays demonstrated that exosomes impair the dif-
3.3. Effects of cancer-derived exosomes on other non-immune
ferentiation of GM-CSF-stimulated BM precursor cells into DCs, as
cells
well as their maturation, which is partially dependent on exosome-
induced IL-6 production [57]. A crucial role for the MyD88-TLR
Apart from endothelium modifications, tumour-derived exo-
signalling pathway in MDSCs activation has also been evaluated
somes may be capable of altering the metastatic niche by directly
[58]. After treatment with tumour exosomes, MyD88 knockout
targeting other non-immune stromal cells, including mesenchymal
mice displayed a less prominent accumulation of MDSCs in pre-
stem cells, fibroblasts and epithelial cells and, in this way, promote
metastatic lung and liver tissues when compared to wild-type
tumour-supportive processes through extracellular matrix (ECM)
animals [58]. This was associated with reduced tumour exosome-
remodelling and induction of angiogenesis. For example, exosomes
promoted lung metastasis, as well as with decreased TNF-a and
produced by human chronic lymphocytic leukemia cells were
IL-6 levels in the serum, and low CCL2 production in the lung
shown to be taken up by mesenchymal stem cells in vitro [47]. As a
[58]. Mechanistically, it seems that the heat shock protein HSP70
result, the cell proliferation of the recipient cells is increased, as is
is involved in tumour exosome-mediated MDSCs induction in
their migration and adhesion to tumour cells [47]. In addition, these
secondary organs, possibly by triggering p-Stat3 in MDSCs in a
recipient mesenchymal stem cells are associated with enhanced
TLR2-MyD88-dependent manner [61].
secretion of inflammatory cytokines that seemed to contribute
More recently, we have shown the tissue-specific immune-
to tumour cell survival [47]. In fact, TGF-␤-containing exosomes
suppressive effects of exosomes derived from breast cancer cells
obtained from various cancer cell lines (including mesothelioma,
with differing metastatic potential [62]. Fluorescently-labelled
prostate and gastric cancer cells) can drive distinct changes in
tumour exosomes injected into syngeneic mice accumulated pre-
primary fibroblasts and mesenchymal stem cells consistent with
dominantly in the lung and were preferentially taken up by CD11b+
their differentiation into a myofibroblast-like phenotype [48,49].
BMDCs, DCs and macrophages, regardless of the metastatic pheno-
This includes high production of FGF2, VEGF, HGF and MMPs, and
type of the cell of origin. Nevertheless, conditioning of tumour naïve
acquisition of both pro-angiogenic and tumour-promoting charac-
animals with exosomes derived from highly metastatic breast can-
teristics [50,51]. The cellular release of soluble factors, such as IL-6
cer cells induced a significant recruitment of MDSCs to lung and
by cholangiocarcinoma-derived exosome-treated BM mesenchy-
liver compared to non-metastatic isogenic exosomes, which was
mal stem cells, also altered tumour cell proliferation, which was
accompanied by a reduced frequency of NK cells [62]. The formation
partially dependent on IL-6R activation [52]. Finally, EMMPRIN,
of this immune-suppressed microenvironment was then associ-
an integral membrane protein shed on lung carcinoma-derived
ated with a higher metastatic burden in both secondary organs,
exosomes, was found to stimulate the production of MMPs by
highlighting that metastatic cancers are highly efficient at prepar-
fibroblasts, outlining another mechanism of tumour-stromal cell
ing a permissive microenvironment that enhances metastatic
interaction that could influence ECM remodelling in the tumour
disease progression [62].
microenvironment [53].
Strikingly, exosome uptake by BMDCs appears to permanently
Recently tumour exosomal small RNA has been demonstrated
alter cellular phenotype. This alteration was shown with a recon-
to be required for neutrophil recruitment to the pre-metastatic
stitution experiment of lethally irradiated control mice with stem
niche after activating resident epithelial cells [54]. This neutrophil
cells from exosome-treated mice that resulted in a significant
recruitment is mediated by the expression of toll like receptor
increase in the number and size of melanoma metastases, as well
(TLR) 3 in alveolar type II lung epithelial cells. Small RNA species
as in the number of metastasis-associated BMDCs [15]. A more
contained within exosomes stimulate activation of TLR3 in lung
detailed assessment of the composition of the bone marrow after
epithelial cells, thereby increasing chemokine production (CXCL1,
reconstitution showed an elevated frequency of pro-angiogenic
CXCL2, CXCL5, and CXCL12) and neutrophil recruitment to pre-
c-kit+ Tie2+ cells after exosome conditioning [15]. Compared to
metastatic sites. Neutrophil recruitment has been shown to be
exosomes derived from poorly metastatic melanoma cells, exo-
involved in cancer metastasis [55,56]. Subsequently, neutrophil
somes from highly metastatic cell lines expressed higher levels
recruitment initiates pro-metastatic inflammation and is related
of metastasis-related proteins, including the MET tyrosine kinase
to the expression of the key metastatic molecules S100A8 and
receptor. The transfer of MET was implicated in the pro-metastatic
S100A9 [54]. This reprogramming of the pre-metastatic niche by
changes of bone marrow progenitors, given the key role of MET
small RNAs in exosomes is conducive to metastatic invasion and
signalling in migration, and angiogenesis [63].
colonisation of secondary organs. These interactions of tumour-
Selective in vivo uptake of pancreatic cancer exosomes by Kupf-
derived exosomes with pre-metastatic niche cells, which in turn
fer cells was also shown to play a key role in liver pre-metastatic
recruit BMDCs indicates that pre-metastatic niche formation is not
niche formation [13]. Kupffer are specialised macrophages localised
simply a binary process, but rather a complicated orchestration of
to the liver, and the transfer of macrophage inhibitory factor (MIF)
signalling involving numerous cells.
from tumour-derived exosomes to these hepatic cells promoted
TFG-␤ production. This generated a fibrotic microenvironment
3.4. Impact of cancer-derived exosomes on immune cells in through increased fibronectin expression from hepatic stellate
pre-metastatic niches cells, as well as further arrest of BM-derived macrophages and neu-
trophils in the liver. These myriad changes ultimately enhanced
Among all the BMDCs subsets, CD11b+ MDSCs are the most metastatic burden, providing key insights into pancreatic cancer
commonly associated with tumour progression and pre-metastatic liver metastasis [13].
niche generation. Exosomes isolated from different types of tumour Additional studies are required to further our understanding
cells (including breast cancer [35,57–59], melanoma [58], thymoma of the effects of tumour exosomes on immune cells functions, as
[59], colon carcinoma [59] and multiple myeloma [60]) have been well as their exact contribution to pre-metastatic niche formation.
shown to modulate BMDCs behaviour, by either directly skewing However, these findings collectively indicate that tumour-derived
their differentiation into MDSCs (as aforementioned) or affecting exosomes promote metastasis by conditioning the BMDCs pheno-
BMDCs homing to pre-metastatic sites. type and/or directly interact with immune cell lineages present in

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
6 R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

the pre-metastatic niche. However, some questions still remain to tropic cancer-cell population into the lung, demonstrating their
be addressed such as: (1) if similar observations can be extended to ability to redirecting metastatic spread of tumour cells [14].
all types of cancer; (2) which immune cell populations are precisely The organotropic nature of exosomes is largely attributed to a
affected by which subsets of circulating exosomes after their target specific repertoire of exosomal surface integrins that was found to
to different organs; (3) the alterations in tissue-specific immune dictate exosome homing to distant organs, as well as uptake by dis-
composition induced by cancer exosome accumulation; (4) which tinct target cells, thereby promoting a favourable pre-metastatic
exosomal content is responsible for driving the opposing effects niche [14]. For instance, ␣6 ␤4 - and ␣6 ␤1 -expressing exosomes
on anti-cancer immune regulation; and (5) the consequences of preferentially interacted with fibroblasts and epithelial cells in the
exosomal content on immune surveillance during metastatic dis- lung, whereas ␣v␤5 integrin-enriched exosomes were preferen-
semination. In this way, expanding our knowledge about how tially taken up by Kupffer cells within the liver microenvironment
exosomes impact immune cells to alter their function and induce a [14]. Perhaps most interesting was the finding that integrins ␤4
tumour-supportive environment may improve the efficacy of cur- and ␣v expression in exosomes isolated from the plasma of can-
rent cancer immunotherapeutics, a highly promising novel class of cer patients could predict metastasis to lung and liver, respectively.
treatment. Interestingly, this finding suggests that circulating tumour-derived
exosomes can be utilised as biomarkers to determine organotropic
metastatic outgrowth [14]. While this work greatly extended our
3.5. Modification of the metabolic environment
understanding of the molecular mechanisms involved in organ-
specific metastasis, and corroborated the role of exosomes on the
In a somewhat unique aspect of pre-metastatic niche biol-
formation of pre-metastatic niche as well as on tumour progression,
ogy, exosomal miRNA has also been demonstrated to reprogram
it also raised several questions. Bone-tropic exosomes, for exam-
the metabolic environment of the pre-metastatic niche. Circulat-
ple, were unable to redirect lung-tropic cell metastasis, and also
ing miR-122 has been associated with metastasis in breast cancer
displayed a limited integrin repertoire [14]. Thus, integrin profiles
patients [64]. Interestingly overexpression of miR-122 in an exper-
alone are unlikely to account for the promotion of organotropic
imental breast cancer model reduced primary tumour growth,
metastasis in all types of cancer, suggesting that additional organ-
yet enhanced metastasis [65]. miR-122 is contained within breast
specific traits of exosomes will still need to be defined. Given these
cancer-derived exosomes and taken up by recipient cells in the
data, it is apparent further investigation is required to determine
pre-metastatic niche of the brain and the lung in vivo [65]. Uptake
how these achievements can be translated to the clinical man-
of miR-122 by recipient cells specifically reduces pyruvate kinase
agement of cancer patients, especially on the use of exosomal
(PKM) expression, and results in decreased GLUT1 and glucose
molecular profiles in both cancer diagnostics and therapeutics.
uptake. When miR-122 containing exosomes were injected into
mice, the expression of PKM and GLUT1 in the brain, and the
lung was reduced. Metabolic reprogramming of the pre-metastatic 3.7. Non-tumour-derived exosomes
niche enhanced metastasis, presumably due to the increased nutri-
ent availability for arriving tumour cells. Cancer cells typically Tumour-derived exosomes are certainly capable of promot-
have a high energy demand, and this unique line of investigation ing metastasis, however it is also apparent that autocrine and
demonstrates that cancer cells can systemically suppress nutrient paracrine signalling at secondary microenvironments – indepen-
utilisation of residing cells in secondary organs in order to favour dent of cancer cells – are integral in the metastatic seeding and
incoming cancer cells. outgrowth of cancer cells [67]. Recently, exosomes derived from
cells residing at secondary microenvironments have been shown
to be essential for metastatic outgrowth after extravasation [67]. In
3.6. Determination of organotropic homing
particular, astrocyte-derived exosomes were shown to be respon-
sible for inducing PTEN loss in brain metastases. PTEN loss was
Organotropic homing of cancer cells to specific organs is of sig-
induced by astrocyte-derived exosomal miR-19a, and was essential
nificant importance in metastatic research. This phenomenon has
for metastatic outgrowth in the brain due to the reciprocal secre-
largely been demonstrated with tumour-derived soluble factors.
tion of CCL2 and recruitment of IBA1-expressing myeloid cells. This
That is, mice treated with melanoma-derived conditioned medium
specific response to the microenvironment of the brain enhanced
redirected metastatic outgrowth of lung carcinoma cells to sites
cancer cell proliferation and reduced apoptosis, highlighting the
typical of melanoma but not lung cancer metastasis [5]. This sug-
importance of understanding not only tumour-derived exosomes,
gested that tumour-derived factors could be capable of inducing
but also exosomes derived from other cell types at secondary
organotropic specific homing. In this context, exosomes have been
organs.
recently identified as drivers of organotropic metastatic spread,
through preferential accumulation at future metastatic sites. This
was first established with melanoma-derived exosomes which 4. Clinical utility of exosomes
accumulated in sentinel lymph nodes, whereas control liposomes
(man-made lipid-based nanoparticles) evenly spread to regional The evident and rapid emergence of numerous publications
and distant nodes [66]. Exosome localisation to these sites was on extracellular vesicles has highlighted the importance of these
accompanied by several modifications in the microenvironment, extracellular components as biomarkers of disease, particularly
such as matrix deposition and vascular proliferation, and thereby cancer. There is currently a large unmet clinical need to develop
facilitated lymphatic melanoma metastasis, implicating exosomes minimally-invasive prognostic or diagnostic markers for most can-
in not only pre-metastatic niche formation, but specific organ- cer types. Given that exosomes contain a myriad of novel proteomic
otropic modulation [66]. Organotropism of exosomes is supported and RNA biomarkers [68,69], which have important functions,
by the assessment of the biodistribution and molecular profiles of particularly in metastatic dissemination and pre-metastatic niche
exosomes obtained from several cancer cells with distinct organ- formation, tumour-derived exosomes are being touted as the next
otropism [14]. Exosomes accumulate in different tissues, however, significant advancement in the biomarker quest [69]. However,
exosome localisation recapitulated the organ-specificity of secret- there are still several hurdles to overcome for retrospective and
ing cells [14]. More strikingly, exosomes obtained from lung-tropic prospective clinical investigations. Technical standardisation is of
breast cancer cells could promote the dissemination of a bone- the upmost importance for utilising exosome biomarkers [70]. We

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 7

have previously demonstrated that different isolation protocols [14] A. Hoshino, B. Costa-Silva, T.-L. Shen, G. Rodrigues, A. Hashimoto, M. Tesic
will deliver significantly different purities, and therefore, con- Mark, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams,
N. Soplop, K. Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, T.
tain various abundances of non-exosomal contaminants that may Zhang, H. Zhang, J. Hernandez, J.M. Weiss, V.D. Dumont-Cole, K. Kramer, L.H.
confound subsequent analysis [12]. Given the disparate isolation Wexler, A. Narendran, G.K. Schwartz, J.H. Healey, P. Sandstrom, K. Jørgen
techniques currently being employed worldwide, a complete stan- Labori, E.H. Kure, P.M. Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur,
M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, M.S. Brady, O. Fodstad, V. Muller,
dardisation of isolation techniques is critical, particularly in order K. Pantel, A.J. Minn, M.J. Bissell, B.A. Garcia, Y. Kang, V.K. Rajasekhar, C.M.
to reproduce retrospective trials, and of course, prospective clin- Ghajar, I. Matei, H. Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins
ical trials, to confirm the validity of the putative exosome-based determine organotropic metastasis, Nature 527 (7578) (2015) 329–335.
[15] H. Peinado, M. Aleckovic, S. Lavotshkin, I. Matei, B. Costa-Silva, G.
biomarker.
Moreno-Bueno, M. Hergueta-Redondo, C. Williams, G. Garcia-Santos, C.M.
Even given these limitations, we are at an exciting stage of Ghajar, A. Nitadori-Hoshino, C. Hoffman, K. Badal, B.A. Garcia, M.K. Callahan, J.
biomarker discovery utilising exosomes. We have discussed the Yuan, V.R. Martins, J. Skog, R.N. Kaplan, M.S. Brady, J.D. Wolchok, P.B.
Chapman, Y. Kang, J. Bromberg, D. Lyden, Melanoma exosomes educate bone
integral role exosomes serve in tumour progression at the primary
marrow progenitor cells toward a pro-metastatic phenotype through MET,
tumour, but also how exosomes are capable of enhancing metas- Nat. Med. 18 (6) (2012) 883–891.
tasis and numerous steps of the metastatic cascade. Exosomes are [16] N. Syn, L. Wang, G. Sethi, J.-P. Thiery, B.-C. Goh, Exosome-mediated
capable of these feats through the specific RNA and protein con- metastasis: from epithelial–mesenchymal transition to escape from
immunosurveillance, Trends Pharmacol. Sci. 37 (7) (2016) 606–617.
tent they carry, and we are now in a unique position to hijack the [17] P. Vaupel, A. Mayer, Hypoxia in cancer: significance and impact on clinical
information carried by exosomes in order to understand, and per- outcome, Cancer Metastasis Rev. 26 (2) (2007) 225–239.
haps interfere with their “intended” purposes, to improve patient [18] M. Hockel, P. Vaupel, Tumor hypoxia: definitions and current clinical,
biologic, and molecular aspects, J. Natl. Cancer Inst. 93 (4) (2001) 266–276.
outcome through diagnostic measures and superior clinical man- [19] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144
agement. (5) (2011) 646–674.
[20] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (1) (2000)
57–70.
Conflict of interest [21] C. Grange, M. Tapparo, F. Collino, L. Vitillo, C. Damasco, M.C. Deregibus, C.
Tetta, B. Bussolati, G. Camussi, Microvesicles released from human renal
The authors declare no conflict of interest. cancer stem cells stimulate angiogenesis and formation of lung premetastatic
niche, Cancer Res. 71 (15) (2011) 5346.
[22] P. Kucharzewska, H.C. Christianson, J.E. Welch, K.J. Svensson, E. Fredlund, M.
Acknowledgements Ringnér, M. Mörgelin, E. Bourseau-Guilmain, J. Bengzon, M. Belting, Exosomes
reflect the hypoxic status of glioma cells and mediate hypoxia-dependent
activation of vascular cells during tumor development, Proc. Natl. Acad. Sci.
The authors would like to thank the members of the Tumour 110 (18) (2013) 7312–7317.
Microenvironment Laboratory for critically reading the manuscript. [23] T. Umezu, H. Tadokoro, K. Azuma, S. Yoshizawa, K. Ohyashiki, J.H. Ohyashiki,
Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances
These authors are supported by the National Health and Medical
angiogenesis by targeting factor-inhibiting HIF-1, Blood 124 (25) (2014)
Research Council Australia (NHMRC, APP1068510) and the Rio- 3748–3757.
Tinto-Ride-To-Conquer-Cancer (6156). [24] C.E. Meacham, S.J. Morrison, Tumour heterogeneity and cancer cell plasticity,
Nature 501 (7467) (2013) 328–337.
[25] K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A. Guha, J. Rak,
References Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles
derived from tumour cells, Nat. Cell Biol. 10 (5) (2008) 619–624.
[1] I.J. Fidler, The pathogenesis of cancer metastasis: the ‘seed and soil’ [26] M. Demory Beckler, J.N. Higginbotham, J.L. Franklin, A.J. Ham, P.J. Halvey, I.E.
hypothesis revisited, Nat. Rev. Cancer 3 (6) (2003) 453–458. Imasuen, C. Whitwell, M. Li, D.C. Liebler, R.J. Coffey, Proteomic analysis of
[2] S. Paget, The distribtuion of secondary growths in cancer of the breast, Lancet exosomes from mutant KRAS colon cancer cells identifies intercellular
133 (3421) (1889) 571–573. transfer of mutant KRAS, Mol. Cell. Proteomics 12 (2) (2013) 343–355.
[3] I.J. Fidler, The pathogenesis of cancer metastasis: the ‘seed and soil’ [27] D. Xiao, S. Barry, D. Kmetz, M. Egger, J. Pan, S.N. Rai, J. Qu, K.M. McMasters, H.
hypothesis revisited, Nat. Rev. Cancer 3 (6) (2003) 453–458. Hao, Melanoma cell-derived exosomes promote epithelial-mesenchymal
[4] G. Poste, I.J. Fidler, The pathogenesis of cancer metastasis, Nature 283 (5743) transition in primary melanocytes through paracrine/autocrine signaling in
(1980) 139–146. the tumor microenvironment, Cancer Lett. 376 (2) (2016) 318–327.
[5] R.N. Kaplan, R.D. Riba, S. Zacharoulis, A.H. Bramley, L. Vincent, C. Costa, D.D. [28] F. Balkwill, A. Mantovani, Inflammation and cancer: back to Virchow? Lancet
MacDonald, D.K. Jin, K. Shido, S.A. Kerns, Z. Zhu, D. Hicklin, Y. Wu, J.L. Port, N. 357 (9255) (2001) 539–545.
Altorki, E.R. Port, D. Ruggero, S.V. Shmelkov, K.K. Jensen, S. Rafii, D. Lyden, [29] G.L. Semenza, P.P. Ruvolo, Introduction to tumor microenvironment
VEGFR1-positive haematopoietic bone marrow progenitors initiate the regulation of cancer cell survival, metastasis, inflammation, and immune
pre-metastatic niche, Nature 438 (7069) (2005) 820–827. surveillance, Biochim. Biophys. Acta 1863 (3) (2016) 379–381.
[6] A.R. Chin, S.E. Wang, Cancer tills the premetastatic field: mechanistic basis [30] M.T. Chow, A. Moller, M.J. Smyth, Inflammation and immune surveillance in
and clinical implications, Clin. Cancer Res. 22 (15) (2016) 3725. cancer, Semin. Cancer Biol. 22 (1) (2012) 23–32.
[7] G.K. Alderton, Metastasis: exosomes drive premetastatic niche formation, [31] T.L. Whiteside, Exosomes and tumor-mediated immune suppression, J. Clin.
Nat. Rev. Cancer 12 (7) (2012), 447-447. Invest. 126 (4) (2016) 1216–1223.
[8] A.S. Azmi, B. Bao, F.H. Sarkar, Exosomes in cancer development, metastasis, [32] J.W. Kim, E. Wieckowski, D.D. Taylor, T.E. Reichert, S. Watkins, T.L. Whiteside,
and drug resistance: a comprehensive review, Cancer Metastasis Rev. 32 (3) Fas ligand-positive membranous vesicles isolated from sera of patients with
(2013) 623–642. oral cancer induce apoptosis of activated T lymphocytes, Clin. Cancer Res.:
[9] B. Gyorgy, T.G. Szabo, M. Pasztoi, Z. Pal, P. Misjak, B. Aradi, V. Laszlo, E. Off. J. Am. Assoc. Cancer Res. 11 (3) (2005) 1010–1020.
Pallinger, E. Pap, A. Kittel, G. Nagy, A. Falus, E.I. Buzas, Membrane vesicles, [33] D.D. Taylor, Ç. Gerçel-Taylor, K.S. Lyons, J. Stanson, T.L. Whiteside, T-Cell
current state-of-the-art: emerging role of extracellular vesicles, Cell Mol. Life apoptosis and suppression of T-Cell receptor/CD3-␨ by fas ligand-containing
Sci. 68 (16) (2011) 2667–2688. membrane vesicles shed from ovarian tumors, Clin. Cancer Res. 9 (14) (2003)
[10] C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and 5113.
function, Nat. Rev. Immunol. 2 (8) (2002) 569–579. [34] A.R. Pyzer, L. Cole, J. Rosenblatt, D.E. Avigan, Myeloid-derived suppressor cells
[11] R. Xu, D.W. Greening, H.J. Zhu, N. Takahashi, R.J. Simpson, Extracellular vesicle as effectors of immune suppression in cancer, Int. J. Cancer 139 (9) (2016)
isolation and characterization: toward clinical application, J. Clin. Invest. 126 1915–1926.
(4) (2016) 1152–1162. [35] X. Xiang, A. Poliakov, C. Liu, Y. Liu, Z.-b. Deng, J. Wang, Z. Cheng, S.V. Shah, G.-J.
[12] R.J. Lobb, M. Becker, S.W. Wen, C.S. Wong, A.P. Wiegmans, A. Leimgruber, A. Wang, L. Zhang, W.E. Grizzle, J. Mobley, H.-G. Zhang, Induction of
Moller, Optimized exosome isolation protocol for cell culture supernatant and myeloid-derived suppressor cells by tumor exosomes, Int. J. Cancer 124 (11)
human plasma, J. Extracell. Vesicles 4 (2015) 27031. (2009) 2621–2633.
[13] B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, A. [36] G. Mignot, F. Chalmin, S. Ladoire, C. Rébé, F. Ghiringhelli, X. Xiang, D.D. Taylor,
Becker, A. Hoshino, M.T. Mark, H. Molina, J. Xiang, T. Zhang, T.-M. Theilen, G. W. Grizzle, H.-G. Zhang, Tumor exosome-mediated MDSC activation, Am. J.
Garcia-Santos, C. Williams, Y. Ararso, Y. Huang, G. Rodrigues, T.-L. Shen, K.J. Pathol. 178 (3) (2011) 1403–1405.
Labori, I.M.B. Lothe, E.H. Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. [37] E.U. Wieckowski, C. Visus, M. Szajnik, M.J. Szczepanski, W.J. Storkus, T.L.
Grandgenett, M.A. Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, Whiteside, Tumor-derived microvesicles promote regulatory t cell expansion
R.E. Schwartz, I. Matei, H. Peinado, B.Z. Stanger, J. Bromberg, D. Lyden, and induce apoptosis in tumor-reactive activated CD8+ t lymphocytes, J.
Pancreatic cancer exosomes initiate pre-metastatic niche formation in the Immunol. 183 (6) (2009) 3720–3730.
liver, Nat. Cell Biol. 17 (6) (2015) 816–826.

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004
G Model
YSCDB-2184; No. of Pages 8 ARTICLE IN PRESS
8 R.J. Lobb et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

[38] P.J. Schuler, Z. Saze, C.S. Hong, L. Muller, D.G. Gillespie, D. Cheng, M. [56] T. El Rayes, R. Catena, S. Lee, M. Stawowczyk, N. Joshi, C. Fischbach, C.A.
Harasymczuk, M. Mandapathil, S. Lang, E.K. Jackson, T.L. Whiteside, Human Powell, A.J. Dannenberg, N.K. Altorki, D. Gao, V. Mittal, Lung inflammation
CD4 + CD39+ regulatory T cells produce adenosine upon co-expression of promotes metastasis through neutrophil protease-mediated degradation of
surface CD73 or contact with CD73+ exosomes or CD73+ cells, Clin. Exp. Tsp-1, Proc. Natl. Acad. Sci. U. S. A. 112 (52) (2015) 16000–16005.
Immunol. 177 (2) (2014) 531–543. [57] S. Yu, C. Liu, K. Su, J. Wang, Y. Liu, L. Zhang, C. Li, Y. Cong, R. Kimberly, W.E.
[39] R. Valenti, V. Huber, P. Filipazzi, L. Pilla, G. Sovena, A. Villa, A. Corbelli, S. Fais, Grizzle, C. Falkson, H.-G. Zhang, Tumor exosomes inhibit differentiation of
G. Parmiani, L. Rivoltini, Human tumor-released microvesicles promote the bone marrow dendritic cells, J. Immunol. 178 (11) (2007) 6867–6875.
differentiation of myeloid cells with transforming growth factor-␤–mediated [58] Y. Liu, X. Xiang, X. Zhuang, S. Zhang, C. Liu, Z. Cheng, S. Michalek, W. Grizzle,
suppressive activity on t lymphocytes, Cancer Res. 66 (18) (2006) 9290. H.-G. Zhang, Contribution of MyD88 to the tumor exosome-mediated
[40] K. Ridder, A. Sevko, J. Heide, M. Dams, A.-K. Rupp, J. Macas, J. Starmann, M. induction of myeloid derived suppressor cells, Am. J. Pathol. 176 (5) (2010)
Tjwa, K.H. Plate, H. Sültmann, P. Altevogt, V. Umansky, S. Momma, 2490–2499.
Extracellular vesicle-mediated transfer of functional RNA in the tumor [59] F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, B. Mélanie, J.-P. Remy-Martin, W.
microenvironment, OncoImmunology 4 (6) (2015) e1008371. Boireau, A. Rouleau, B. Simon, D. Lanneau, A. De Thonel, G. Multhoff, A.
[41] G.P. Gupta, J. Massague, Cancer metastasis: building a framework, Cell 127 (4) Hamman, F. Martin, B. Chauffert, E. Solary, L. Zitvogel, C. Garrido, B. Ryffel, C.
(2006) 679–695. Borg, L. Apetoh, C. Rébé, F. Ghiringhelli, Membrane-associated Hsp72 from
[42] S. Hiratsuka, K. Nakamura, S. Iwai, M. Murakami, T. Itoh, H. Kijima, J.M. tumor-derived exosomes mediates STAT3-dependent immunosuppressive
Shipley, R.M. Senior, M. Shibuya, MMP9 induction by vascular endothelial function of mouse and human myeloid-derived suppressor cells, J. Clin.
growth factor receptor-1 is involved in lung-specific metastasis, Cancer Cell 2 Invest. 120 (2) (2010) 457–471.
(4) (2002) 289–300. [60] J. Wang, K. De Veirman, S. Faict, M.A. Frassanito, D. Ribatti, A. Vacca, E. Menu,
[43] J. Sceneay, M.T. Chow, A. Chen, H.M. Halse, C.S.F. Wong, D.M. Andrews, E.K. Multiple myeloma exosomes establish a favourable bone marrow
Sloan, B.S. Parker, D.D. Bowtell, M.J. Smyth, A. Möller, Primary tumor hypoxia microenvironment with enhanced angiogenesis and immunosuppression, J.
recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises Pathol. 239 (2) (2016) 162–173.
NK cell cytotoxicity in the premetastatic niche, Cancer Res. 72 (16) (2012) [61] J. Diao, X. Yang, X. Song, S. Chen, Y. He, Q. Wang, G. Chen, C. Luo, X. Wu, Y.
3906. Zhang, Exosomal Hsp70 mediates immunosuppressive activity of the
[44] H. Peinado, S. Lavotshkin, D. Lyden, The secreted factors responsible for myeloid-derived suppressor cells via phosphorylation of Stat3, Med. Oncol.
pre-metastatic niche formation: old sayings and new thoughts, Semin. Cancer 32 (2) (2015) 1–10.
Biol. 21 (2) (2011) 139–146. [62] S.W. Wen, J. Sceneay, L.G. Lima, C.S. Wong, M. Becker, S. Krumeich, R.J. Lobb,
[45] J. Sceneay, M.J. Smyth, A. Möller, The pre-metastatic niche: finding common V. Castillo, K.N. Wong, S. Ellis, The biodistribution and immune suppressive
ground, Cancer Metastasis Rev. 32 (3) (2013) 449–464. effects of breast cancer-derived exosomes, Cancer Res. 76 (23) (2016)
[46] W. Zhou, M.Y. Fong, Y. Min, G. Somlo, L. Liu, M.R. Palomares, Y. Yu, A. Chow, 6816–6827.
S.T. O’Connor, A.R. Chin, Y. Yen, Y. Wang, E.G. Marcusson, P. Chu, J. Wu, X. Wu, [63] L. Trusolino, A. Bertotti, P.M. Comoglio, MET signalling: principles and
A.X. Li, Z. Li, H. Gao, X. Ren, M.P. Boldin, P.C. Lin, S.E. Wang, Cancer-secreted functions in development, organ regeneration and cancer, Nat. Rev. Mol. Cell
miR-105 destroys vascular endothelial barriers to promote metastasis, Cancer Biol. 11 (12) (2010) 834–848.
Cell 25 (4) (2014) 501–515. [64] X. Wu, G. Somlo, Y. Yu, M.R. Palomares, A.X. Li, W. Zhou, A. Chow, Y. Yen, J.J.
[47] J. Paggetti, F. Haderk, M. Seiffert, B. Janji, U. Distler, W. Ammerlaan, Y.J. Kim, J. Rossi, H. Gao, J. Wang, Y.C. Yuan, P. Frankel, S. Li, K.T. Ashing-Giwa, G. Sun, Y.
Adam, P. Lichter, E. Solary, G. Berchem, E. Moussay, Exosomes released by Wang, R. Smith, K. Robinson, X. Ren, S.E. Wang, De novo sequencing of
chronic lymphocytic leukemia cells induce the transition of stromal cells into circulating miRNAs identifies novel markers predicting clinical outcome of
cancer-associated fibroblasts, Blood 126 (9) (2015) 1106. locally advanced breast cancer, J. Transl. Med. 10 (2012) 42.
[48] J. Webber, R. Steadman, M.D. Mason, Z. Tabi, A. Clayton, Cancer exosomes [65] M.Y. Fong, W. Zhou, L. Liu, A.Y. Alontaga, M. Chandra, J. Ashby, A. Chow, S.T.
trigger fibroblast to myofibroblast differentiation, Cancer Res. 70 (23) (2010) O’Connor, S. Li, A.R. Chin, G. Somlo, M. Palomares, Z. Li, J.R. Tremblay, A.
9621. Tsuyada, G. Sun, M.A. Reid, X. Wu, P. Swiderski, X. Ren, Y. Shi, M. Kong, W.
[49] J. Gu, H. Qian, L. Shen, X. Zhang, W. Zhu, L. Huang, Y. Yan, F. Mao, C. Zhao, Y. Zhong, Y. Chen, S.E. Wang, Breast-cancer-secreted miR-122 reprograms
Shi, W. Xu, Gastric cancer exosomes trigger differentiation of umbilical cord glucose metabolism in premetastatic niche to promote metastasis, Nat. Cell
derived mesenchymal stem cells to carcinoma-associated fibroblasts through Biol. 17 (2) (2015) 183–194.
TGF-␤/Smad pathway, PLoS One 7 (12) (2012) e52465. [66] J.L. Hood, R.S. San, S.A. Wickline, Exosomes released by melanoma cells
[50] R. Chowdhury, J.P. Webber, M. Gurney, M.D. Mason, Z. Tabi, A. Clayton, Cancer prepare sentinel lymph nodes for tumor metastasis, Cancer Res. 71 (11)
exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic (2011) 3792.
and pro-invasive myofibroblasts, Oncotarget 6 (No 2) (2015). [67] L. Zhang, S. Zhang, J. Yao, F.J. Lowery, Q. Zhang, W.C. Huang, P. Li, M. Li, X.
[51] J.P. Webber, L.K. Spary, A.J. Sanders, R. Chowdhury, W.G. Jiang, R. Steadman, J. Wang, C. Zhang, H. Wang, K. Ellis, M. Cheerathodi, J.H. McCarty, D. Palmieri, J.
Wymant, A.T. Jones, H. Kynaston, M.D. Mason, Z. Tabi, A. Clayton, Saunus, S. Lakhani, S. Huang, A.A. Sahin, K.D. Aldape, P.S. Steeg, D. Yu,
Differentiation of tumour-promoting stromal myofibroblasts by cancer Microenvironment-induced PTEN loss by exosomal microRNA primes brain
exosomes, Oncogene 34 (3) (2015) 290–302. metastasis outgrowth, Nature 527 (7576) (2015) 100–104.
[52] H. Haga, I.K. Yan, K. Takahashi, J. Wood, A. Zubair, T. Patel, Tumour cellderived [68] A. Thind, C. Wilson, Exosomal miRNAs as cancer biomarkers and therapeutic
extracellular vesicles interact with mesenchymal stem cells to modulate the targets, J. Extracell. Vesicles 5 (2016) 31292.
microenvironment and enhance cholangiocarcinoma growth, J. Extracell. [69] T. An, S. Qin, Y. Xu, Y. Tang, Y. Huang, B. Situ, J.M. Inal, L. Zheng, Exosomes
Vesicles 4 (2015), incl supplements. serve as tumour markers for personalized diagnostics owing to their
[53] S.S. Sidhu, A.T. Mengistab, A.N. Tauscher, J. LaVail, C. Basbaum, The important role in cancer metastasis, J. Extracell. Vesicles 4 (2015) 27522.
microvesicle as a vehicle for EMMPRIN in tumor-stromal interactions, [70] K.W. Witwer, E.I. Buzas, L.T. Bemis, A. Bora, C. Lasser, J. Lotvall, E.N. Nolte-’t
Oncogene 23 (4) (2004) 956–963. Hoen, M.G. Piper, S. Sivaraman, J. Skog, C. Thery, M.H. Wauben, F. Hochberg,
[54] Y. Liu, Y. Gu, Y. Han, Q. Zhang, Z. Jiang, X. Zhang, B. Huang, X. Xu, J. Zheng, X. Standardization of sample collection, isolation and analysis methods in
Cao, Tumor exosomal RNAs promote lung pre-metastatic niche formation by extracellular vesicle research, J. Extracell. Vesicles 2 (2013).
activating alveolar epithelial TLR3 to recruit neutrophils, Cancer Cell 30 (2)
(2016) 243–256.
[55] J. Cools-Lartigue, J. Spicer, B. McDonald, S. Gowing, S. Chow, B. Giannias, F.
Bourdeau, P. Kubes, L. Ferri, Neutrophil extracellular traps sequester
circulating tumor cells and promote metastasis, J. Clin. Invest. 123 (8) (2013)
3446–3458.

Please cite this article in press as: R.J. Lobb, et al., Exosomes: Key mediators of metastasis and pre-metastatic niche formation, Semin
Cell Dev Biol (2017), http://dx.doi.org/10.1016/j.semcdb.2017.01.004

You might also like