You are on page 1of 9

Advanced Drug Delivery Reviews 106 (2016) 148156

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

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

Extracellular vesicles for drug delivery


Pieter Vader a,, Emma A. Mol b, Gerard Pasterkamp a,b, Raymond M. Schiffelers a,
a
Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
b
Department of Experimental Cardiology, University Medical Center Utrecht, the Netherlands, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

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

Article history: Extracellular vesicles (EVs) are cell-derived membrane vesicles, and represent an endogenous mechanism for in-
Received 7 January 2016 tercellular communication. Since the discovery that EVs are capable of functionally transferring biological infor-
Received in revised form 11 February 2016 mation, the potential use of EVs as drug delivery vehicles has gained considerable scientic interest. EVs may
Accepted 17 February 2016
have multiple advantages over currently available drug delivery vehicles, such as their ability to overcome natu-
Available online 27 February 2016
ral barriers, their intrinsic cell targeting properties, and stability in the circulation. However, therapeutic applica-
Keywords:
tions of EVs as drug delivery systems have been limited due to a lack of methods for scalable EV isolation and
Extracellular vesicles efcient drug loading. Furthermore, in order to achieve targeted drug delivery, their intrinsic cell targeting prop-
exosomes erties should be tuned through EV engineering. Here, we review and discuss recent progress and remaining chal-
microvesicles lenges in the development of EVs as drug delivery vehicles.
drug delivery 2016 Elsevier B.V. All rights reserved.
isolation
biodistribution
targeting
nanomedicine

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
2. Isolation of extracellular vesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
3. Loading of extracellular vesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
3.1. Loading of cells before extracellular vesicle isolation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
3.2. Loading of extracellular vesicles after isolation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
4. Biodistribution and targeting of extracellular vesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
4.1. Circulation time and biodistribution of extracellular vesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
4.2. Interactions of extracellular vesicles with the immune system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
4.3. Specic targeting of extracellular vesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153
5. Therapeutic effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153
6. Extracellular vesicle-mimetic nanovesicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Conict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155

1. Introduction
This review is part of the Advanced Drug Delivery Reviews theme issue on Biologically-
inspired drug delivery systems. Extracellular vesicles (EVs) are nano-sized membrane vesicles,
Corresponding authors at: Department of Clinical Chemistry and Hematology ,
University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
released by many, if not all, cell types. EV release has been found to
E-mail addresses: pvader@umcutrecht.nl (P. Vader), r.schiffelers@umcutrecht.nl occur in many unicellular- as well as multicellular organisms, suggest-
(R.M. Schiffelers). ing that it represents an evolutionary-conserved process. Mammalian

http://dx.doi.org/10.1016/j.addr.2016.02.006
0169-409X/ 2016 Elsevier B.V. All rights reserved.
P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156 149

cells can release distinct types of EVs, including exosomes, microvesicles, 2. Isolation of extracellular vesicles
and apoptotic bodies. This classication is based on their intracellular or-
igin. Exosomes are thought to be the smallest vesicle type, with sizes One of the prerequisites for clinical application of EVs is standardiza-
ranging from 40 to 100 nm. They originate from intraluminal budding tion of the isolation process with regards to yield, reproducibility, and
of multivesicular endosomes (MVE) and are released upon fusion of purity [25]. Furthermore, such an isolation method should be scalable
MVEs with the plasma membrane. In contrast, microvesicles (also and robust. For large scale EV production, the manufacturing process se-
referred to as ectosomes) are more heterogeneous in size and can quentially involves expansion of the donor cell line, collection of the
be larger (50 nm-1 m), and are formed through direct budding conditioned medium, and EV purication. Thus far, several methods
from the plasma membrane. When cells are compelled to undergo for the isolation of EVs have been described. The most commonly used
apoptosis, a heterogeneous population of vesicles (50 nm-5 m) is method is differential ultracentrifugation (UC). EVs are isolated based
released, which are named apoptotic bodies [1]. Despite these differ- on sedimentation at high g-forces. Generally, this method comprises
ences in origin, no uniform EV nomenclature exists as of yet, due to low speed spins to remove cell debris, followed by high speed spins to
the overlap in vesicle sizes and the absence of subtype-specic pellet EVs. Sucrose density gradients may subsequently be utilized to
markers. As a result, it remains difcult (if not impossible) to purify separate vesicle types based on density, and to further purify vesicles
and thereby distinguish between vesicle types [2]. In this review, from protein aggregates [26,27]. Disadvantages of UC and sucrose gradi-
we therefore refer to all vesicle subtypes as extracellular vesicles ents include the time-consuming protocol, operator-dependent yield,
(EVs). and possible aggregation and rupture of EVs due to high shear forces
The cargo of EVs comprises small and long, coding and non-coding [28]. Isolation of EVs using UC may therefore not be useful for clinical
RNAs (e.g. mRNA, miRNA, lncRNA), lipids and proteins [3,4]. Initially, practice, and novel isolation techniques are topic of intensive investiga-
EVs were thought to act as garbage bags, with a main function in tion. Two distinct approaches for isolation can be discriminated.
discard of cellular waste. Over the last decade, however, scientic The rst approach, immunoafnity isolation, is based on selective
interest in EVs has rapidly increased, after it was shown that biolog- capture of EVs that bear specic marker proteins on their surface. This
ical information packaged in EVs could be transferred between cells, could be important when separation of EV subtypes is required, al-
and alter the recipient cells phenotype. Cells can package a distinct though it is currently unknown whether specic subtypes are more or
set of biomolecules into EVs via endogenous sorting mechanisms, less feasible for drug delivery purposes. Clayton et al. developed an iso-
and release EVs constitutively or after stimulation [5,6]. EVs may lation method to capture EVs derived from antigen-presenting cells
subsequently be internalized by target recipient cells, resulting in (APCs) using antibody-coated magnetic beads. Using antibodies specic
transfer of mRNAs and miRNAs, which can result in production or for major histocompatibility complex (MHC) class II, a specic EV sub-
silencing of target proteins, respectively [79], and of proteins, in- type (i.e. exosomes) could be isolated [29]. A different antibody-based
cluding membrane proteins [10,11]. EVs can be isolated from bodily method to isolate EVs was described by Ashcroft et al., who used a
uids, including blood, urine, cerebrospinal uid and saliva [1214]. microuidic circuit to isolate CD41-positive platelet-derived EVs in plas-
As their content reects the status of the donor cell, EVs may be ap- ma. EVs were captured with an anti-CD41 antibody-coated mica surface
plied in diagnostics, either as pathological biomarkers or to follow [30]. This standardized and quick method requires a very low amount of
treatment efcacy (reviewed in [15,16]). In addition, it has become plasma (10 l) and could be adjusted for other sources of EVs in the fu-
clear that, through their important role in intercellular communica- ture. However, the absence of well-dened EV markers may thus lead to
tion, EVs affect various processes involved in health and disease. The isolation of only specic EV subsets or EVs derived from specic cell
discovery that EVs make up a natural mechanism for information types, and successful elution of intact EVs from the beads might prove
transfer between cells has stimulated interest into their potential challenging. Furthermore, immunoafnity isolation protocols are not
use as a new drug delivery platform. very attractive for clinical applications, since EVs are isolated at a very
Despite considerable research in the last 50 years, the clinical small scale.
translation of conventional drug delivery platforms has been limited. The second approach comprises methods that isolate EVs based on
The efciency of these platforms to overcome barriers in macromol- their size. With these methods, considerable efforts have already been
ecule drug transport, such as reaching the target tissue and engaging made to improve scalability of EV isolation. Lamparski et al. showed in-
intracellular targets, is still unsatisfactory [17]. In addition, concerns creased recovery of MHC class II-expressing EVs using a combination of
related to immunogenicity and toxicity of non-natural delivery ultraltration and ultracentrifugation of EVs in a 30% sucrose/deuterium
systems remain. EVs on the other hand seem to have many features oxide cushion, for the rst time showing that it is possible to isolate EVs
of an ideal carrier system. The EV cargo is naturally protected from for clinical application [31]. However, a drawback of this method is the
degradation in the circulation [18]. EVs seem to possess intrinsic low EV recovery, hindering application in large clinical studies. In an at-
cell targeting properties, and are able to overcome natural barriers tempt to isolate EVs using ltration techniques only, Heinemann et al.
such as the blood-brain barrier [19,20]. Furthermore, it is likely developed an easy three-step protocol [32]. First, a 0.1 m pore size pol-
that EVs utilize endogenous mechanisms for uptake, intracellular yethersulfone (PES) membrane was used to remove dead cells and cell
trafcking and subsequent delivery of their content in recipient debris. The sample was then passed through a 500 kDa molecular
cells [21]. Importantly, EVs may be nearly non-immunogenic when weight cut-off modied PES lter to remove free proteins and reduce
used autologously. Moreover, several clinical trials using EVs for im- large volumes, followed by isolation of EVs using a 0.1 um Track Etch l-
munotherapy have already demonstrated the safety of EV adminis- ter. A comparison of sequential ltration versus UC showed that al-
tration in humans [2224]. though ltration resulted in a slight reduction of EV yield compared to
Although EVs hold immense promise for therapeutic drug delivery, UC, it resulted in isolation of a more specic subset of EVs. The major ad-
clinical applications may critically depend on the development of vantage of this method is the fast and fully automatable protocol, al-
scalable EV isolation techniques and approaches for efcient drug though non-specic EV protein binding to the membranes leading to
loading. Furthermore, improved methods to modify their in vivo lower recovery may present a limitation.
biodistribution, which is an important determinant of their thera- The use of size-exclusion chromatography (SEC) for EV isolation
peutic effect as it enables more specic drug delivery to target tis- from plasma was rst described by Boing et al. [33]. Fractionation of
sues, are required. In this review, we discuss new ndings and plasma using a sepharose CL-2B column resulted in fast and specic
recent improvements on these issues, and summarize recent suc- separation of proteins, HDL, and EVs. SEC isolation also resulted in a
cesses in the use of EVs as drug delivery vehicles in animal disease higher recovery of EVs compared to UC. Increased standardization of
models. EV isolation was reported by Welton et al., who employed a
150 P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156

commercially available column [34]. Furthermore, collection of only the mechanisms that are involved in RNA sorting into EVs largely remain
EV-containing eluate fractions reduced the isolation time to 10-15 mi- to be elucidated [5,6,42].
nutes. Recently, Nordin et al. reported the isolation of EVs from large Although most studies to date have investigated small RNAs as ther-
volumes of cell culture media by ultraltration combined with size- apeutic cargo to be loaded in EVs, other types of therapeutics may also
exclusion chromatography (SEC) [35]. The sample volume was rst re- be incorporated, including mRNAs, proteins, and small molecules.
duced by ultraltration using 100 kDa-cutoff spin lters, followed by EV Pascucci et al. recently showed successful loading of Paclitaxel (PTX)
isolation using SEC. They demonstrated that this method resulted in a into EVs, by incubating mesenchymal stromal cells (MSCs) with a high
higher EV yield when compared to UC. Furthermore, electron microsco- dosage of PTX. Subsequently, MSCs produced EVs loaded with PTX,
py data showed more biophysically intact EVs after SEC isolation, which which were shown to be able to inhibit in vitro tumor growth [43]. In ad-
had impact on their subsequent biodistribution in vivo. These novel dition, Tang et al. showed that tumor cells incubated with chemothera-
methods based on ltration and/or SEC could very well be adjusted peutic drugs package these drug into EVs. To stimulate formation of
and optimized for large-scale clinical application, including for drug de- drug-loaded EVs, cells were irradiated with ultraviolet light to induce
livery purposes. apoptosis [44]. Lee et al. recently demonstrated for a variety of com-
Taken together, these studies also underline that medical application pounds that delivery into cells using fusogenic liposomes also leads to
of EVs still poses signicant challenges. From a pharmaceutical perspec- their loading into EVs released from these cells [45]. Although shown
tive, the reproducibility of composition and purity is particularly de- to be feasible for loading of hydrophobic as well as hydrophilic com-
manding. Especially since we know that out of the different classes of pounds, it is unclear whether this approach also leads to incorporation
biomolecules associated with EVs, such as nucleic acids, proteins and of synthetic lipids from the liposomes into EVs.
lipids, there are hundreds of species of each class that may contribute
to the overall effects. In many instances, the therapeutic molecules 3.2. Loading of extracellular vesicles after isolation
have only partly been identied. This is important as identication of
the active agents in the composition dictates quality control. It is con- The second approach involves loading of EVs after their isolation. For
ceivable that, similar as for stem cell therapy, a framework of Good Prac- some hydrophobic drugs, EV loading may be achieved through direct
tices will need to be established in which also the particular mixing. This approach was employed by Sun et al., who loaded EVs
pharmacokinetic and pharmacodynamic qualities of the EV product with curcumin, an anti-inammatory drug, by simple incubation at
are dened on a case-by-case basis [25]. At the same time, the rst clin- 22C for 5 minutes. Retention in EVs increased curcumin in vitro solubil-
ical studies have already been conducted demonstrating that clinical ap- ity and stability, and in vivo bioavailability. Intraperitoneal injection of
plication of EVs is safe and feasible. curcumin-loaded EVs resulted in protection against lipopolysaccharide
(LPS)-induced septic shock in mice, suggesting that this loading ap-
proach may also be feasible for clinical use [46]. Mixing has also been
3. Loading of extracellular vesicles
successfully employed to load PTX into EVs [47]. Others have shown
that mild sonication may further improve PTX incorporation [48].
The lipid bilayer membrane serves as a natural barrier to protect EV
For hydrophilic compounds, including RNA, a major obstacle is
cargo from degradation in the bloodstream. However, the presence of
formed by the lipid bilayer membrane, which restricts passive loading
this membrane as well as the EV endogenous content make loading of
into EVs. One suggested method to achieve small RNA loading after EV
exogenous cargo (i.e. therapeutics) into EVs challenging. Nevertheless,
isolation is electroporation. This approach is based on spontaneous
several methods to load EVs have already been described. Here, we pro-
pore formation in membranes to compensate for changes in voltage
vide an overview of two different loading approaches. One approach is
after stimulation with an electrical signal. Alvarez-Erviti et al. were the
based on loading of therapeutics into cells from which the EVs are de-
rst to report apparent successful siRNA loading into EVs by electropo-
rived, which, using the endogenous loading machinery of the cells,
ration. Importantly, subsequent systemic administration of siRNA-
may result in subsequent EV loading with the drug of interest. The sec-
loaded EVs in mice resulted in inhibition of Beta-Site APP-Cleaving En-
ond approach involves loading of EVs after their isolation. An overview
zyme 1 mRNA and protein expression in the brain [19]. The use of elec-
of the described loading techniques is schematically depicted in Fig. 1.
troporation for siRNA loading was also investigated by Walhgren et al.
Encapsulation was conrmed by uorescent microscopy, northern blot-
3.1. Loading of cells before extracellular vesicle isolation ting, and ow cytometry. EV-mediated delivery of siRNA to monocytes
and lymphocytes was observed, which resulted in inhibition of
EVs are natural carriers of small RNAs, including miRNAs. Binding of mitogen-activated protein kinase 1 in vitro [49]. These studies suggested
miRNAs (or siRNAs) to a target mRNA can cause either mRNA degrada- that electroporation could lead to encapsulation of small RNAs in EVs,
tion or repression of protein translation, resulting in target gene silenc- without affecting EV integrity and function. In contrast, when
ing. This process is known as RNA interference [36]. Small RNAs may Kooijmans et al. investigated the efciency of electroporation for
also be used therapeutically, for the treatment of diseases in which spe- siRNA loading into EVs, no signicant differences in siRNA retention be-
cic genes are overactive, such as cancer. However, RNAs typically re- tween samples electroporated in the presence or absence of EVs could
quire delivery to their site-of-action, which is the cytosol of target be measured [50]. Instead, they found that electroporation of siRNA re-
cells. To achieve loading of small RNAs into EVs, transfection-based ap- sults in formation of extensive siRNA aggregates even in the absence of
proaches have been proposed. Kosaka et al. showed that transfection of EVs, which could lead to possible overestimation of the siRNA encapsu-
HEK293 and COS-7 cells with a vector for expression of miR-16, -21, lation efciency. Therefore, caution should be taken with the interpreta-
-143, -146a, or -155 resulted in overexpression of these miRNAs in tion of studies using electroporation for loading in the absence of proper
cells and, as a result of endogenous RNA secretory mechanisms, in control experiments. In addition, electroporation might cause aggrega-
their subsequent active release in EVs [37]. Subsequently, these tion or fusion of EVs themselves, as has been shown for liposomes
miRNA-containing EVs were able to induce gene silencing in recipient [51]. In an attempt to minimize EV aggregation after electroporation,
COS-7 cells. Similarly, other reports have shown that using vector- Hood et al. investigated the use of membrane stabilizers. The use of tre-
induced expression of small RNAs in cells, small RNA loading into EVs halose pulse media (TPM) was shown to maximize EV colloidal stability,
can be achieved [38,39]. Alternatively, EV donor cells may be transfected most likely due to the biologic stabilization properties of trehalose [52].
with small RNAs directly [40,41]. The main disadvantage of these ap- In an attempt to load EVs with the antioxidant enzyme catalase,
proaches however is that the level of RNA that is incorporated into several loading strategies were investigated by Haney et al., includ-
EVs may depend on the RNA species and/or sequence, although the ing simple incubation at room temperature, saponin-mediated
P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156 151

Fig. 1. Animated overview of extracellular vesicle loading strategies. Left panel: Loading of EVs via transfection of the donor cell with a vector encoding small RNA or via incubation of
the donor cell with small molecules. Using the endogenous sorting machinery of donor cell, EVs are subsequently loaded with the therapeutics. Right panels: Loading of EVs after their
isolation. Electroporation is based on spontaneous pore formation after stimulation with an electrical signal. Other approaches include incubation at RT, repeated freeze-thaw cycles,
application of ultrasonic frequencies, treatment with a detergent-like molecule (e.g. saponin), or extrusion. Abbreviations: EV = extracellular vesicle, RT = room temperature.

permeabilization, sonication, freeze-thaw cycles, and extrusion [53]. possible disruption of the EV integrity during sonication or extrusion
The highest loading efciencies were obtained with sonication, extru- procedures or saponin treatment affects their immunogenicity. A simi-
sion or saponin treatment, although sonication and extrusion of EVs re- lar comparison of strategies for loading of hydrophilic porphyrins was
sulted in signicant size increases as observed by dynamic light performed by Fuhrmann et al [54]. They observed an eleven-fold in-
scattering, nanoparticle tracking analysis and atomic force microscopy. crease in loading efcacy after EV loading by saponin treatment com-
EVs loaded by sonication or by saponin treatment protected neurons pared to loading by passive incubation or extrusion. They also
against reactive oxygen species production in vitro and, after intranasal compared EV loading with hydrophobic and hydrophilic porphyrins
administration, decreased microglial activity compared to injection of by passive incubation at room temperature or electroporation. EV load-
free catalase or PBS in an acute brain inammation mouse model. Al- ing efciency was higher for hydrophobic porphyrins compared to the
though this study demonstrates the feasibility of these strategies for hydrophilic compounds. Interestingly, in both cases electroporation in-
loading EV with large proteins, it remains to be determined how creased the encapsulation efciency compared to passive incubation.
152 P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156

Hydrophobic porphyrin-loaded EVs showed improved cellular uptake similar to that of phosphatidylcholine : cholesterol liposomes and lipo-
compared to free drug or porphyrins-loaded liposomes. Reduced cell vi- somes made from the lipid extract of EVs [60]. Further studies should re-
ability was observed in cancer cells after treatment with porphyrin- veal whether EV clearance by the MPS is in fact due to their intrinsic
loaded EVs followed by irradiation. Together, these studies demonstrate properties, or whether it is a result of the exogenous characteristics of
that various approaches for EV loading are starting to become available. EVs obtained from in vitro cultures, presence of non-self-labels that
Still, selection of the most suitable method for clinical application re- are present on the EV surface, or the quantity of vesicles that are
mains to be determined. injected [59]. Interestingly, Kooijmans et al. recently showed that pro-
viding stealth properties to EVs via introduction of polyethylene glycol
4. Biodistribution and targeting of extracellular vesicles (PEGylation) onto the EV surface signicantly increased their circula-
tion time in mice. PEGylation of EVs may therefore be employed to in-
4.1. Circulation time and biodistribution of extracellular vesicles crease accumulation in tumors or at sites of inammation, which are
characterized by localized increased vascular permeability [61].
In vivo biodistribution and targeting of specic tissues by ther- Besides reports describing biodistribution of EVs after systemic in-
apeutics are important factors for their effectiveness. Issues associ- jection, the possibility of EV administration via other routes has also
ated with administration of free drugs, including unfavorable been investigated. For some therapeutic applications, local drug admin-
pharmacokinetics and lack of selectivity, may be overcome by the istration may actually be preferred, e.g. when targeting the central ner-
use of delivery vehicles. For this reason, Wiklander et al. investi- vous system. Zhuang et al. showed that intranasal administration of
gated the tissue distribution of 1,1'-dioctadecyl-3,3,3',3'- mouse lymphoma EVs loaded with the anti-inammatory drug
tetramethylindotricarbocyanine iodide (DiR)-labeled EVs from curcumin resulted in their localization to the brain. Curcumin levels
various cell sources [55]. Twenty-four hours after intravenous peaked at 1 hour after administration, and a signicant amount could
(i.v.) injection in mice, they observed the highest uorescent sig- still be detected after 12 hours. No toxicity or abnormal behavior was
nal in the liver, followed by spleen, gastrointestinal tract, and observed in mice upon EV treatment [20]. In addition, intranasal admin-
lungs. Furthermore, they showed that cell source, EV dose, and istration of catalase-loaded EVs resulted in higher EV accumulation in
route of administration affect EV distribution, which may have im- brain tissue after 4 hours in a mouse model of Parkinsons disease, com-
plications for the design and feasibility of therapeutic studies using pared to intravenous injection [53]. Establishing the most efcient EV
EVs. Injection of higher EV doses resulted in relatively lower liver administration route for each application may therefore help to improve
accumulation compared to lower doses, possibly caused by satura- therapeutic efcacy.
tion of the mononuclear phagocyte system. Systematic comparison
of the biodistribution of intraperitoneally (i.p), subcutaneously (s.c), 4.2. Interactions of extracellular vesicles with the immune system
and i.v. injected EVs revealed that i.p. and s.c. injections resulted in re-
duced EV accumulation in liver and spleen and enhanced pancreas EVs are predominantly submicron particles, which gives them a
and GI tract accumulation compared to i.v. injections. large surface area to volume ratio. In addition, because of their particu-
With a similar aim of visualizing EV biodistribution and clearance late nature, they can also display repeating molecular patterns on their
in vivo, Lai et al. developed an elegant EV imaging reporter system surface which can be important in their interactions with the immune
[56]. EVs were engineered to express a fusion of membrane-bound system. Together, these EV qualities constitute a reactive surface for
Gaussia luciferase (Gluc) reporter and a biotin domain, allowing multi- molecules and cells, especially after injection into the blood stream.
modal imaging. In a time frame of 30 minutes to 6 hours after i.v. injec- Within plasma, several protein cascades might interact with the ve-
tion of Gluc-EVs in athymic nude mice, highest bioluminescent signals sicular surface. Classically, phosphatidylserine present on the extracel-
were observed in spleen and liver. Surprisingly, after subsequent lular vesicle surface has been described to recruit several
transcardial perfusion with PBS, highest Gluc-EV signals were observed phosphatidylserine-binding proteins of the coagulation cascade. The
in kidneys, and perfused spleens showed only minimal signals across all vesicles provide a catalytic surface by bringing the proteins in close
time points. This difference suggests that the majority of splenic accu- proximity to each other, thereby increasing reaction speed. As a result,
mulation is caused by EV storage in the spleen rather than uptake by EVs are generally described to promote coagulation and thrombosis.
the tissue itself. Comparable Gluc-EV signals before and after perfusion Another important protein cascade in plasma is the complement
were observed for liver and lungs, thus indicating active uptake of EVs in system. Complement has been described to be activated by liposomes
these organs. Most EVs were cleared after 6 hours, as a result of active and contribute to their clearance [62]. For EVs, the situation seems to
uptake of EVs by cells or hepatic clearance. Imai et al. employed a similar be more complex. For human antigen presenting cells, for example,
approach to assess the importance of macrophages in EV clearance. EVs contain CD55 and CD59 membrane regulators of complement.
After systemic administration of Gluc-lactadherin (Gluc-LA)-labeled Both regulators were shown to be important in limiting complement
EVs derived from melanoma cells, macrophage-depleted mice deposition on the vesicles as well as EV lysis [63]. These results indicate
displayed higher Gluc-LA EV accumulation in liver, spleen, and lungs, that EVs carry complement inhibitors that may promote their survival
and delayed EV clearance from blood compared to untreated mice in the blood stream. At the same time, another study on tumor cell de-
[57]. This suggests an important role for macrophages in EV clearance. rived EVs showed that these activated both C5a and soluble S protein
Instead of following EV biodistribution using labeled EVs, Bala et al. bound C5b-9, in a concentration dependent manner [64]. Activation
loaded miRNA-155, an inammatory mediator, into EVs and deter- was dependent on the presence of calcium indicating that activation oc-
mined organ distribution of miRNA-155-loaded EVs after i.v. adminis- curred through the classical or lectin pathway. Interestingly, EVs from
tration in miRNA-155-/- mice. After perfusion, highest miRNA-155 malignant cell lines were more potent in complement activation than
signals were again observed in the liver, followed by adipose tissue EVs from a non-malignant cell line. Taken together these results indicate
and lungs, whereas the lowest signals were observed in muscle and kid- that the source and composition of EVs is important in their net effect on
neys [58]. the complement cascade.
Together, these reports demonstrate that systemically administered The intrinsic tissue distribution prole of isolated EVs underlines
EVs (1) have a short half-life and (2) are rapidly taken up by the mono- that a large fraction of EVs interacts with the RES. In particular macro-
nuclear phagocyte system (MPS), particularly in the liver and spleen. phages in liver and spleen are responsible for EV uptake, which could
Seemingly, this mechanism of clearance resembles that described for impact the cellular phenotype. This intrinsic uptake by antigen-
synthetic nanoparticles, such as liposomes [59]. Moreover, it was re- presenting cells has been regarded as a possible route for vaccination.
cently shown that the EV clearance rate and biodistribution prole is Because of the resemblance of EV composition to the composition of
P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156 153

the parental cell, EVs can contain antigens, for example oncogenes. As eld gradient, represents a non-invasive alternative strategy to enhance
such, the EVs could represent a platform for tumor vaccination. Al- therapeutic efcacy. Delivery of drugs to tissues using magnetic iron
though some studies support dendritic cell- and tumor-derived EVs as oxide nanoparticles bound to a therapeutic agent was already intro-
anticancer vaccines (reviewed in [65]), recent literature suggests that duced in the 1970s [73,74]. Recently, Silva et al. provided proof-of-
EVs are predominantly immunosuppressive [6668]. It is likely that concept of using the combination of EVs and magnetic targeting for
the condition of the immune system and the state and composition of tissue-specic delivery. Therapeutics and iron oxide nanoparticles
the parental cell dictates the outcome of the encounter of immune sys- were together incubated with macrophages, which resulted in produc-
tem and EVs. An important pathophysiological role of this interaction is tion of EVs loaded with both the therapeutic and magnetic nanoparti-
illustrated in a recent paper by Costa-Silva et al. [69]. Here they showed cles. Magnetic targeting resulted in enhanced as well as spatially
that uptake of pancreatic carcinoma-derived exosomes by Kupffer cells modulated EV and drug uptake by cancer cells in vitro [75]. However,
in the liver caused these cells to secrete transforming growth factor- disadvantages of this method when applied in vivo may include difcul-
beta and hepatic stellate cells to produce bronectin. This environment ties to target deep tissues in the body, and toxicity issues associated
promoted inux of bone marrow-derived macrophages that secreted with the use of iron nanoparticles.
macrophage migration inhibitory factor that was shown to be involved
in liver metastasis of the pancreatic cancer cells. This study underlines 5. Therapeutic effects
the important consequences of interaction of EVs with components of
the immune system. Despite the challenges in the isolation, loading and targeting of EVs,
several reports have already demonstrated their potential for thera-
4.3. Specic targeting of extracellular vesicles peutic drug delivery in various animal models of disease. The major-
ity of these studies have focussed on cancer therapy. Due to their
As described above, administration of exogenous EVs seems to result nanosize, EVs may achieve passive targeting to tumors via the en-
mainly in accumulation in the spleen and liver. However, several studies hanced permeation and retention (EPR) effect [76]. One of the rst
have shown that it is possible to engineer EVs in such a way, that in- studies that described the use of EVs for drug delivery to tumors
creased targeting to other tissues or specic cell types is achieved. A was performed by Mizrak et al. EVs derived from the cell line HEK-
well-studied strategy to equip EVs with targeting properties is via trans- 293 were loaded with the suicide gene cytosine deaminase fused to
fection of EV-producing cells to drive expression of targeting moieties uracil phosphoribosyltransferase to induce cell death in nerve sheath
fused with EV membrane proteins. To accomplish increased targeting tumor cells upon addition of the prodrug 5-uorocytosine. Weekly
to the brain, Alvarez-Erviti et al. engineered dendritic cells (DCs) to ex- intratumoral injections of engineered EVs led to signicant tumor re-
press a fusion protein of an EV membrane anchor, lysosome-associated gression in mice after 2 months [77]. Later, Ohno et al. showed that
membrane glycoprotein 2b (Lamp2b), and brain-specic rabies viral EGFR- targeted EVs loaded with let-7a miRNA delivered this miRNA
glycoprotein (RVG), a peptide that binds the acetylcholine receptor. to EGFR-expressing breast tumors upon systemic delivery in
EVs isolated from these cells were shown to express RVG on their sur- RAG2-/- mice, which signicantly inhibited tumor development
face. After tail vein injection of siRNA-loaded EVs, knockdown of [72]. However, downregulation of let-7a target genes could not be
BACE1 mRNA and protein was demonstrated in the brains of mice, for demonstrated. In addition, Zhang et al. utilized EVs to deliver
the rst time showing the potential of EVs as targeted drug delivery sys- siRNA against transforming growth factor 1 (TGF1), a well-
tems [19]. Wiklander et al. indeed found increased accumulation of studied target for cancer treatment, to sarcoma cells [78]. Using a
these RVG-targeted EVs in the brain after systemic administration com- subcutaneous tumor model in mice, they showed that i.v. injection
pared to unmodied EVs [55]. Similarly, Tian et al. showed that expres- of TGF-1 siRNA-loaded EVs strongly suppressed TGF-1 expression
sion of a fusion of Lamp2b and v integrin-specic iRGD peptide and downstream signaling in the tumors, and inhibited primary
(internalizing RGD peptide CRGDKGPDC) in immature DCs resulted in tumor growth as well as formation of lung metastases. Interestingly,
expression of these peptides on the surface of EVs. Injection of modied siRNA doses used in this study were surprisingly low (0.4 pmol per
EVs loaded with the chemotherapeutic drug doxorubicin in tumor- injection). This may indicate that EVs were highly efcient in deliv-
bearing mice resulted in delivery of doxorubicin to the tumor site [70]. ering their cargo to the target cells, or that other EV-mediated effects
However, Hung & Leonard found that peptides fused to the N- contributed to the observed inhibition of tumor growth.
terminus of Lamp2b are not detected in cells and on EVs, presumably Besides for delivery of small RNA molecules, which critically depend
as a result of degradation by endosomal proteases [71]. Glycosylation on intracellular delivery to conduct their silencing function, EVs have
of these peptides protected against proteolytic degradation, which en- also been employed to deliver chemotherapeutic agents, with the aim
hanced expression on EVs and increased target binding. Subsequent to to enhance their efcacy and reduce side effects. For example, Tian
the study of Alvarez-Erviti et al., they also investigated the inuence of et al. observed signicantly improved suppression of breast tumor
engineered glycosylation on the uptake of RVG-displaying EVs by neu- growth after i.v. injection of integrin-targeted, dendritic cell-derived
roblastoma cells. EVs expressing glycosylated RVG peptides fused to EVs loaded with the chemotherapeutic drug doxorubicin in mice, com-
Lamp2b were more efciently internalized in vitro compared to EVs ex- pared to free drug. Moreover, doxorubicin was shown to cause less car-
pressing non-glycosylated peptides. Therefore, glycosylation of diac damage, the most important dose-limiting side effect of the drug,
targeting peptides could possibly enhance tissue targeting. when packaged in EVs [70]. Furthermore, PTX-loaded EVs were shown
To achieve targeted delivery to epidermal growth factor receptor to be more effective for inhibiting growth of Lewis lung carcinoma me-
(EGFR)-expressing tumors, Ohno et al. displayed EGFR-specic GE11- tastases than Taxol, a commercially available formulation of PTX [48].
peptides fused to transmembrane domains of platelet-derived growth Tang et al. used two different tumor models to show benecial effects
factor receptor on EVs. They showed that miRNA-loaded EVs derived of packaging chemotherapeutic drugs into EVs [44]. Repeated i.p. injec-
from the human embryonic kidney cell line HEK293 modied to ex- tions of cisplatin-loaded EVs improved long-term survival of ovarian
press the GE11-peptide displayed increased accumulation in EGFR- cancer-bearing mice, as compared to cisplatin only. Furthermore, i.v. in-
expressing breast tumors in mice compared to untargeted EVs [72]. jection of doxorubicin-loaded EVs delayed growth of established subcu-
Together, these data highlight the possibilities of engineering of EVs to taneous hepatocarcinoma. Importantly, these EV treatments did not
enhance specic tissue targeting, which could contribute to making adversely affect liver or kidney functions, which are typical side effects
EVs applicable as drug delivery systems. observed after administration of the free drugs. Based on results from
Besides ligand-mediated targeting, magnetic drug targeting, i.e. en- this study (Fig. 2), a phase II clinical trial has been initiated which
hanced drug delivery to a chosen tissue by application of a magnetic aims to evaluate the effect of chemotherapeutic drugs encapsulated in
154 P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156

Fig. 2. Schematic overview of preclinical studies that formed the basis for the rst clinical trials using extracellular vesicles for drug delivery. Left panel: Loading of the anti-
inammatory agent curcumin into EVs was achieved via simple incubation at 22C. Curcumin-loaded EVs were shown to attenuate autoimmune encephalitis and LPS-induced septic
shock in mice. Right panel: EVs loaded with chemotherapeutic drugs inhibited hepatocarcinoma in mice after tail vein injection. EV loading was achieved by rst incubating tumor
cells with chemotherapeutic drugs. Then, to stimulate formation of drug-loaded EVs, cells were irradiated with ultraviolet light to induce apoptosis. Abbreviations: EV = extracellular
vesicle, LPS = lipopolysaccharide, i.p. = intraperitoneal, UV = ultraviolet.

EVs on malignant ascites and pleural effusion in advanced cancer 6. Extracellular vesicle-mimetic nanovesicles
patients. Patients are locally injected four times a week, and both
therapeutic as well as side effects are recorded (clinicaltrials.gov, Although substantial progress has been made in the development of
NCT01854866). natural EVs as drug delivery vehicles, the relatively low recovery of EVs
A second ongoing clinical trial (phase I) in which EVs are being eval- produced by mammalian cells remains an obstacle for large-scale EV
uated as drug delivery systems is based on two studies that showed production. For this reason, the possibility of generating EV-mimetic
benecial effects of encapsulating anti-inammatory compounds into vesicles with a substantially greater yield has attracted recent attention.
EVs (Fig. 2). Firstly, Sun et al. showed that EVs as vehicles for curcumin Methods for preparing EV-mimetics are based on obtaining articially
increase its solubility, stability, and bioavailability. Furthermore, generated nanovesicles from broken cells, which resemble the structur-
curcumin-loaded EVs protected mice from lipopolysaccharide (LPS)- al and physical features of EVs. For example, Jang et al. produced drug-
induced septic shock [46]. In a follow-up study, daily intranasal admin- loaded nanovesicles by serial extrusion of monocytes through lters in
istrations of curcumin packaged in EVs delayed and attenuated experi- the presence of chemotherapeutics, and isolated these vesicles from
mental autoimmune encephalomyelitis, an effect not observed after free drugs using an OptiPrep density gradient [79]. This production
administration of curcumin alone. Mechanistically, treatment success method resulted in a 100-fold increased vesicle yield compared to isola-
was likely caused by increased induction of apoptosis in microglial tion of naturally produced EVs. Nanovesicles were shown to express
cells [20]. The clinical trial aims to study the ability of plant EVs to lymphocyte function-associated antigen-1 (LFA-1) that can bind endo-
deliver curcumin to colon tumors. Outcome measures include thelial cell adhesion molecules (CAMs), overexpressed on activated en-
curcumin concentrations in normal and cancerous tissue after oral dothelial cells, such as found in tumors. Importantly, i.v. injection of
administration and safety as well as tolerability (compared to these drug-loaded nanovesicles in mice resulted in their accumulation
curcumin alone) as determined by adverse events (clinicaltrials. in tumor tissue and subsequent reduction of tumor growth, without
gov, NCT01294072). the toxic side effects present after injection of the free drug. Interesting-
Besides for cancer, EVs have been proposed as therapeutic delivery ly, EV-mimetic nanovesicles were found to be similarly effective as EVs
vehicle for the treatment of Parkinsons disease (PD). PD is associated harvested from the same cells. An alternative method to generate EV-
with reduced levels of several brain enzymes such as catalase, super- mimetic nanovesicles was proposed by Yoon et al. Living cells were
oxide dismutase, and other antioxidants. To increase brain levels of sliced with microfabricated silicon nitride blades of 500 nm while
catalase, Haney et al. intranasally administered catalase-loaded EVs owing through a standardized microuidic system. After slicing,
in a mouse model of PD [53]. Catalase-loaded EVs were shown to re- nanovesicles were spontaneously formed through self-assembly of
duce microglial activation and protect neurons against reactive oxy- membrane fragments. Vesicles were shown to be composed of a lipid
gen species more efciently compared to free catalase. Altogether, bilayer membrane and carry various membrane proteins, intracellular
these results, although preliminary, have demonstrated that EVs proteins and RNAs. The addition of polystyrene latex beads during slic-
are promising candidate drug carriers for the treatment of a variety ing resulted in encapsulation of beads in the nanovesicles, with efcien-
of diseases. cies up to 30% [80]. These successful examples demonstrate the
P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156 155

potential of these methods to generate nanovesicles for drug delivery evidence for horizontal transfer of mRNA and protein delivery, Leukemia 20
(2006) 847856.
on a large scale. However, whether these cell-derived EV-mimetic vesi- [9] D.M. Pegtel, K. Cosmopoulos, D.A. Thorley-Lawson, M.A. van Eijndhoven, E.S.
cles also possess the envisioned advantages of natural EVs, including Hopmans, J.L. Lindenberg, T.D. de Gruijl, T. Wurdinger, J.M. Middeldorp, Functional
low immunogenicity and intrinsic capacity to utilize endogenous mech- delivery of viral miRNAs via exosomes, Proc. Natl. Acad. Sci. U. S. A. 107 (2010)
63286333.
anisms for uptake and intracellular trafcking in recipient cells, is cur- [10] J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves, W.T.
rently unknown. Curry Jr., B.S. Carter, A.M. Krichevsky, X.O. Breakeeld, Glioblastoma microvesicles
transport RNA and proteins that promote tumour growth and provide diagnostic
biomarkers, Nat. Cell Biol. 10 (2008) 14701476.
7. Conclusions [11] K. Al-Nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May, A. Guha, J. Rak, Intercellular
transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour
EVs are natural membrane vesicles involved in intercellular commu- cells, Nat. Cell Biol. 10 (2008) 619624.
[12] M.P. Caby, D. Lankar, C. Vincendeau-Scherrer, G. Raposo, C. Bonnerot, Exosomal-like
nication. The ability of EVs to transfer their content to recipient cells via
vesicles are present in human blood plasma, Int. Immunol. 17 (2005) 879887.
endogenous uptake mechanisms makes them attractive candidates for [13] T. Pisitkun, R.F. Shen, M.A. Knepper, Identication and proteomic proling of
application in drug delivery. However, current obstacles to overcome exosomes in human urine, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 1336813373.
before deployment of EVs in large clinical trials are the need for both [14] Y. Ogawa, Y. Miura, A. Harazono, M. Kanai-Azuma, Y. Akimoto, H. Kawakami, T.
Yamaguchi, T. Toda, T. Endo, M. Tsubuki, R. Yanoshita, Proteomic Analysis of Two
scalable EV isolation methods, and more efcient drug loading ap- Types of Exosomes in Human Whole Saliva, Biol. Pharm. Bull. 34 (2011) 1323.
proaches for different therapeutics. Especially ultraltration- and SEC- [15] L. Urbanelli, S. Buratta, K. Sagini, G. Ferrara, M. Lanni, C. Emiliani, Exosome-based
based EV isolation approaches seem promising for large-scale clinical strategies for Diagnosis and Therapy, Recent patents on CNS drug discovery,
102015 1027.
application, although further optimizations remain to be implemented. [16] C. Lasser, Exosomes in diagnostic and therapeutic applications: biomarker, vaccine
An additional challenge remains shifting in vivo biodistribution of EVs and RNA interference delivery vehicle, Expert. Opin. Biol. Ther. 15 (2015) 103117.
from non-specic organ accumulation to accumulation in desired tis- [17] Time to deliver, Nat. Biotechnol. 32 (2014) 961.
[18] N.B.Y. Tsui, E.K.O. Ng, Y.M.D. Lo, Stability of endogenous and added RNA in blood
sues. Although considerable efforts have been made in targeting specic specimens, serum, and plasma, Clin. Chem. 48 (2002) 16471653.
cell types by engineering EVs to express cell-type specic ligands, one of [19] L. Alvarez-Erviti, Y.Q. Seow, H.F. Yin, C. Betts, S. Lakhal, M.J.A. Wood, Delivery of
the major obstacles remains inefcient drug delivery to target tissues. siRNA to the mouse brain by systemic injection of targeted exosomes, Nat.
Biotechnol. 29 (2011) (341-U179).
Gaining more insight into fundamental EV biology, in particular into [20] X.Y. Zhuang, X.Y. Xiang, W. Grizzle, D.M. Sun, S.Q. Zhang, R.C. Axtell, S.W. Ju, J.Y. Mu,
how EVs target specic cell types, and whether different EV subtypes L.F. Zhang, L. Steinman, D. Miller, H.G. Zhang, Treatment of Brain Inammatory Dis-
serve different physiological roles, could therefore contribute to further eases by Delivering Exosome Encapsulated Anti-inammatory Drugs From the
Nasal Region to the Brain, Mol. Ther. 19 (2011) 17691779.
improvements in the development of EVs for drug delivery.
[21] M.E. Marcus, J.N. Leonard, FedExosomes: Engineering Therapeutic Biological Nano-
particles that Truly Deliver, Pharmaceuticals (Basel) 6 (2013) 659680.
Conict of interest statement [22] B. Escudier, T. Dorval, N. Chaput, F. Andre, M.P. Caby, S. Novault, C. Flament, C.
Leboulaire, C. Borg, S. Amigorena, C. Boccaccio, C. Bonnerot, O. Dhellin, M.
Movassagh, S. Piperno, C. Robert, V. Serra, N. Valente, J.B. Le Pecq, A. Spatz, O.
RMS is the CSO of Excytex, and on the scientic advisory board of JSR Lantz, T. Tursz, E. Angevin, L. Zitvogel, Vaccination of metastatic melanoma patients
Micro NV. with autologous dendritic cell (DC) derived-exosomes: results of therst phase I
clinical trial, J. Transl. Med. 3 (2005) 10.
[23] M.A. Morse, J. Garst, T. Osada, S. Khan, A. Hobeika, T.M. Clay, N. Valente, R.
Acknowledgements Shreeniwas, M.A. Sutton, A. Delcayre, D.H. Hsu, J.B. Le Pecq, H.K. Lyerly, A phase I
study of dexosome immunotherapy in patients with advanced non-small cell lung
PV is supported by a VENI Fellowship (# 13667) from the cancer, J. Transl. Med. 3 (2005) 9.
[24] S. Dai, D. Wei, Z. Wu, X. Zhou, X. Wei, H. Huang, G. Li, Phase I clinical trial of autol-
Netherlands Organisation for Scientic Research (NWO). The work of ogous ascites-derived exosomes combined with GM-CSF for colorectal cancer, Mol.
EAM is part of the Project SMARTCARE-II of the BioMedical Materials in- Ther. 16 (2008) 782790.
stitute, co-funded by the Dutch Ministry of Economic Affairs, Agricul- [25] T. Lener, M. Gimona, L. Aigner, V. Borger, E. Buza s, G. Camussi, N. Chaput, D.
Chatterjee, F.A. Court, H.A. Del Portillo, L. O'Driscoll, S. Fais, J.M. Falcon-Perez, U.
ture and Innovation and the Netherlands CardioVascular Research Felderhoff-Mueser, L. Fraile, Y.S. Gho, A. Gorgens, R.C. Gupta, A. Hendrix, D.M.
Initiative (CVON): the Dutch Heart Foundation, Dutch Federation of Hermann, A.F. Hill, F. Hochberg, P.A. Horn, D. de Kleijn, L. Kordelas, B.W. Kramer,
University Medical Centers, the Netherlands Organization for Health E.M. Kramer-Albers, S. Laner-Plamberger, S. Laitinen, T. Leonardi, M.J. Lorenowicz,
S.K. Lim, J. Lotvall, C.A. Maguire, A. Marcilla, I. Nazarenko, T. Ochiya, T. Patel, S.
Research and Development, and the Royal Netherlands Academy of
Pedersen, G. Pocsfalvi, S. Pluchino, P. Quesenberry, I.G. Reischl, F.J. Rivera, R.
Sciences. The work of RMS on cell-derived membrane vesicles is sup- Sanzenbacher, K. Schallmoser, I. Slaper-Cortenbach, D. Strunk, T. Tonn, P. Vader,
ported by a European Research Council starting grant (# 260627) B.W. van Balkom, M. Wauben, S.E. Andaloussi, C. Thery, E. Rohde, B. Giebel, Applying
extracellular vesicles based therapeutics in clinical trials - an ISEV position paper, J.
MINDS in the FP7 ideas program of the European Union.
Extracellular Vesicles 4 (2015) 30087.
[26] C. Thery, S. Amigorena, G. Raposo, A. Clayton, Isolation and characterization of
References exosomes from cell culture supernatants and biological uids, Curr. Protoc. Cell
Biol. (2006) 22 (Chapter 3, Unit 3).
[1] M. Colombo, G. Raposo, C. Thery, Biogenesis, secretion, and intercellular interactions [27] M. Aalberts, F.M. van Dissel-Emiliani, N.P. van Adrichem, M. van Wijnen, M.H.
of exosomes and other extracellular vesicles, Annu. Rev. Cell Dev. Biol. 30 (2014) Wauben, T.A. Stout, W. Stoorvogel, Identication of distinct populations of
255289. prostasomes that differentially express prostate stem cell antigen, annexin A1,
[2] S.J. Gould, G. Raposo, As we wait: coping with an imperfect nomenclature for extra- and GLIPR2 in humans, Biol. Reprod. 86 (2012) 82.
cellular vesicles, J. Extracellular Vesicles 2 (2013). [28] D.D. Taylor, S. Shah, Methods of isolating extracellular vesicles impact down-stream
[3] G. Raposo, W. Stoorvogel, Extracellular vesicles: Exosomes, microvesicles, and analyses of their cargoes, Methods 87 (2015) 310.
friends, J. Cell Biol. 200 (2013) 373383. [29] A. Clayton, J. Court, H. Navabi, M. Adams, M.D. Mason, J.A. Hobot, G.R. Newman, B.
[4] S.M. van Dommelen, P. Vader, S. Lakhal, S.A.A. Kooijmans, W.W. van Solinge, M.J.A. Jasani, Analysis of antigen presenting cell derived exosomes, based on immuno-
Wood, R.M. Schiffelers, Microvesicles and exosomes: Opportunities for cell- magnetic isolation and ow cytometry, J. Immunol. Methods 247 (2001) 163174.
derived membrane vesicles in drug delivery, J. Control. Release 161 (2012) 635644. [30] B.A. Ashcroft, J. de Sonneville, Y. Yuana, S. Osanto, R. Bertina, M.E. Kuil, T.H.
[5] D. Koppers-Lalic, M. Hackenberg, I.V. Bijnsdorp, M.A.J. van Eijndhoven, P. Sadek, D. Oosterkamp, Determination of the size distribution of blood microparticles directly
Sie, N. Zini, J.M. Middeldorp, B. Ylstra, R.X. de Menezes, T. Wurdinger, G.A. Meijer, in plasma using atomic force microscopy and microuidics, Biomed. Microdevices
D.M. Pegtel, Nontemplated Nucleotide Additions Distinguish the Small RNA Compo- 14 (2012) 641649.
sition in Cells from Exosomes, Cell Rep. 8 (2014) 16491658. [31] H.G. Lamparski, A. Metha-Damani, J.Y. Yao, S. Patel, D.H. Hsu, C. Ruegg, J.B. Le Pecq,
[6] M.F. Bolukbasi, A. Mizrak, G.B. Ozdener, S. Madlener, T. Strobel, E.P. Erkan, J.B. Fan, Production and characterization of clinical grade exosomes derived from dendritic
X.O. Breakeeld, O. Saydam, miR-1289 and "Zipcode"-like Sequence Enrich cells, J. Immunol. Methods 270 (2002) 211226.
mRNAs in Microvesicles, Mol. Ther.Nucleic Acids 1 (2012). [32] M.L. Heinemann, M. Ilmer, L.P. Silva, D.H. Hawke, A. Recio, M.A. Vorontsova, E. Alt, J.
[7] H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall, Exosome- Vykoukal, Benchtop isolation and characterization of functional exosomes by se-
mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic ex- quential ltration, J. Chromatogr. A 1371 (2014) 125135.
change between cells, Nat. Cell Biol. 9 (2007) 654659. [33] A.N. Boing, E. van der Pol, A.E. Grootemaat, F.A. Coumans, A. Sturk, R. Nieuwland,
[8] J. Ratajczak, K. Miekus, M. Kucia, J. Zhang, R. Reca, P. Dvorak, M.Z. Ratajczak, Embry- Single-step isolation of extracellular vesicles by size-exclusion chromatography, J.
onic stem cell-derived microvesicles reprogram hematopoietic progenitors: Extracellular Vesicles 3 (2014).
156 P. Vader et al. / Advanced Drug Delivery Reviews 106 (2016) 148156

[34] J.L. Welton, J.P. Webber, L.A. Botos, M. Jones, A. Clayton, Ready-made chromatogra- [56] C.P. Lai, O. Mardini, M. Ericsson, S. Prabhakar, C.A. Maguire, J.W. Chen, B.A. Tannous,
phy columns for extracellular vesicle isolation from plasma, J. Extracellular Vesicles X.O. Breakeeld, Dynamic biodistribution of extracellular vesicles in vivo using a
4 (2015) 27269. multimodal imaging reporter, ACS Nano 8 (2014) 483494.
[35] J.Z. Nordin, Y. Lee, P. Vader, I. Mager, H.J. Johansson, W. Heusermann, O.P. Wiklander, [57] T. Imai, Y. Takahashi, M. Nishikawa, K. Kato, M. Morishita, T. Yamashita, A.
M. Hallbrink, Y. Seow, J.J. Bultema, J. Gilthorpe, T. Davies, P.J. Fairchild, S. Gabrielsson, Matsumoto, C. Charoenviriyakul, Y. Takakura, Macrophage-dependent clearance of
N.C. Meisner-Kober, J. Lehtio, C.I. Smith, M.J. Wood, S. El Andaloussi, Ultraltration systemically administered B16BL6-derived exosomes from the blood circulation in
with size-exclusion liquid chromatography for high yield isolation of extracellular mice, J. Extracellular Vesicles 4 (2015) 26238.
vesicles preserving intact biophysical and functional properties, Nanomed.: [58] S. Bala, T. Csak, F. Momen-Heravi, D. Lippai, K. Kodys, D. Catalano, A. Satishchandran,
Nanotechnol., Biol. Med. 11 (4) (May 2015) 879883. V. Ambros, G. Szabo, Biodistribution and function of extracellular miRNA-155 in
[36] S.M. Elbashir, J. Harborth, W. Lendeckel, A. Yalcin, K. Weber, T. Tuschl, Duplexes of mice, Sci. Rep. 5 (2015) 10721.
21-nucleotide RNAs mediate RNA interference in cultured mammalian cells, Nature [59] R. van der Meel, M.H.A.M. Fens, P. Vader, W.W. van Solinge, O. Eniola-Adefeso, R.M.
411 (2001) 494498. Schiffelers, Extracellular vesicles as drug delivery systems: Lessons from the lipo-
[37] N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki, T. Ochiya, Secretory mech- some eld, J. Control. Release 195 (2014) 7285.
anisms and intercellular transfer of microRNAs in living cells, J. Biol. Chem. 285 [60] T. Smyth, M. Kullberg, N. Malik, P. Smith-Jones, M.W. Graner, T.J. Anchordoquy,
(2010) 1744217452. Biodistribution and delivery efciency of unmodied tumor-derived exosomes, J.
[38] N. Kosaka, H. Iguchi, Y. Yoshioka, K. Hagiwara, F. Takeshita, T. Ochiya, Competitive Control. Release 199 (2015) 145155.
interactions of cancer cells and normal cells via secretory microRNAs, J. Biol. [61] S.A. Kooijmans, L.A. Fliervoet, R. van der Meel, M.H. Fens, H.F. Heijnen, P.M. van
Chem. 287 (2012) 13971405. Bergen En Henegouwen, P. Vader, R.M. Schiffelers, PEGylated and targeted extracel-
[39] Q. Pan, V. Ramakrishnaiah, S. Henry, S. Fouraschen, P.E. de Ruiter, J. Kwekkeboom, lular vesicles display enhanced cell specicity and circulation time, J. Control. Re-
H.W. Tilanus, H.L. Janssen, L.J. van der Laan, Hepatic cell-to-cell transmission of lease 224 (2016) 7785.
small silencing RNA can extend the therapeutic reach of RNA interference (RNAi), [62] S.M. Moghimi, I. Hamad, Liposome-mediated triggering of complement cascade, J.
Gut 61 (2012) 13301339. Liposome Res. 18 (2008) 195209.
[40] Y. Zhang, D. Liu, X. Chen, J. Li, L. Li, Z. Bian, F. Sun, J. Lu, Y. Yin, X. Cai, Q. Sun, K. Wang, [63] A. Clayton, C.L. Harris, J. Court, M.D. Mason, B.P. Morgan, Antigen-presenting cell
Y. Ba, Q. Wang, D. Wang, J. Yang, P. Liu, T. Xu, Q. Yan, J. Zhang, K. Zen, C.Y. Zhang, Se- exosomes are protected from complement-mediated lysis by expression of CD55
creted monocytic miR-150 enhances targeted endothelial cell migration, Mol. Cell and CD59, Eur. J. Immunol. 33 (2003) 522531.
39 (2010) 133144. [64] B. Whitehead, L. Wu, M.L. Hvam, H. Aslan, M. Dong, L. Dyrskjot, M.S. Ostenfeld, S.M.
[41] Y. Akao, A. Iio, T. Itoh, S. Noguchi, Y. Itoh, Y. Ohtsuki, T. Naoe, Microvesicle-mediated Moghimi, K.A. Howard, Tumour exosomes display differential mechanical and com-
RNA molecule delivery system using monocytes/macrophages, Mol. Ther. 19 (2011) plement activation properties dependent on malignant state: implications in endo-
395399. thelial leakiness, J. Extracellular Vesicles 4 (2015) 29685.
[42] A.O. Batagov, V.A. Kuznetsov, I.V. Kurochkin, Identication of nucleotide patterns [65] N. Chaput, J. Taieb, F. Andre, L. Zitvogel, The potential of exosomes in immunother-
enriched in secreted RNAs as putative cis-acting elements targeting them to apy, Expert. Opin. Biol. Ther. 5 (2005) 737747.
exosome nano-vesicles, BMC Genomics 12 (Suppl. 3) (2011) S18. [66] J.E. Hellwinkel, J.S. Redzic, T.A. Harland, D. Gunaydin, T.J. Anchordoquy, M.W. Graner,
[43] L. Pascucci, V. Cocce, A. Bonomi, D. Ami, P. Ceccarelli, E. Ciusani, L. Vigano, A. Glioma-derived extracellular vesicles selectively suppress immune responses,
Locatelli, F. Sisto, S.M. Doglia, E. Parati, M.E. Bernardo, M. Muraca, G. Alessandri, G. Neuro Oncol. (Sep. 18 2015) [Epub ahead of print].
Bondiolotti, A. Pessina, Paclitaxel is incorporated by mesenchymal stromal cells [67] M. Monguio-Tortajada, R. Lauzurica-Valdemoros, F.E. Borras, Tolerance in organ
and released in exosomes that inhibit in vitro tumor growth: A new approach for transplantation: from conventional immunosuppression to extracellular vesicles,
drug delivery, J. Control. Release 192 (2014) 262270. Front. Immunol. 5 (2014) 416.
[44] K. Tang, Y. Zhang, H.F. Zhang, P.W. Xu, J. Liu, J.W. Ma, M. Lv, D.P. Li, F. Katirai, G.X. [68] L. Kordelas, V. Rebmann, A.K. Ludwig, S. Radtke, J. Ruesing, T.R. Doeppner, M. Epple,
Shen, G.M. Zhang, Z.H. Feng, D.Y. Ye, B. Huang, Delivery of chemotherapeutic P.A. Horn, D.W. Beelen, B. Giebel, MSC-derived exosomes: a novel tool to treat
drugs in tumour cell-derived microparticles, Nat. Commun. 3 (2012). therapy-refractory graft-versus-host disease, Leukemia 28 (2014) 970973.
[45] J. Lee, J. Kim, M. Jeong, H. Lee, U. Goh, H. Kim, B. Kim, J.H. Park, Liposome-Based En- [69] B. Costa-Silva, N.M. Aiello, A.J. Ocean, S. Singh, H. Zhang, B.K. Thakur, A. Becker, A.
gineering of Cells To Package Hydrophobic Compounds in Membrane Vesicles for Hoshino, M.T. Mark, H. Molina, J. Xiang, T. Zhang, T.M. Theilen, G. Garcia-Santos, C.
Tumor Penetration, Nano Lett. 15 (2015) 29382944. Williams, Y. Ararso, Y. Huang, G. Rodrigues, T.L. Shen, K.J. Labori, I.M. Lothe, E.H.
[46] D. Sun, X. Zhuang, X. Xiang, Y. Liu, S. Zhang, C. Liu, S. Barnes, W. Grizzle, D. Miller, Kure, J. Hernandez, A. Doussot, S.H. Ebbesen, P.M. Grandgenett, M.A.
H.G. Zhang, A novel nanoparticle drug delivery system: the anti-inammatory activ- Hollingsworth, M. Jain, K. Mallya, S.K. Batra, W.R. Jarnagin, R.E. Schwartz, I. Matei,
ity of curcumin is enhanced when encapsulated in exosomes, Mol. Ther. 18 (2010) H. Peinado, B.Z. Stanger, J. Bromberg, D. Lyden, Pancreatic cancer exosomes initiate
16061614. pre-metastatic niche formation in the liver, Nat. Cell Biol. 17 (2015) 816826.
[47] H. Saari, E. Lazaro-Ibanez, T. Viitala, E. Vuorimaa-Laukkanen, P. Siljander, M. [70] Y. Tian, S. Li, J. Song, T. Ji, M. Zhu, G.J. Anderson, J. Wei, G. Nie, A doxorubicin delivery
Yliperttula, Microvesicle- and exosome-mediated drug delivery enhances the cyto- platform using engineered natural membrane vesicle exosomes for targeted tumor
toxicity of Paclitaxel in autologous prostate cancer cells, J. Control. Release 220 therapy, Biomaterials 35 (2014) 23832390.
(2015) 727737. [71] M.E. Hung, J.N. Leonard, Stabilization of exosome-targeting peptides via engineered
[48] M.S. Kim, M.J. Haney, Y. Zhao, V. Mahajan, I. Deygen, N.L. Klyachko, E. Inskoe, A. glycosylation, J. Biol. Chem. 290 (2015) 81668172.
Piroyan, M. Sokolsky, O. Okolie, S.D. Hingtgen, A.V. Kabanov, E.V. Batrakova, Devel- [72] S. Ohno, M. Takanashi, K. Sudo, S. Ueda, A. Ishikawa, N. Matsuyama, K. Fujita, T.
opment of exosome-encapsulated paclitaxel to overcome MDR in cancer cells, Mizutani, T. Ohgi, T. Ochiya, N. Gotoh, M. Kuroda, Systemically injected exosomes
Nanomed.: Nanotechnol., Biol. Med. (Nov. 14 2015), http://dx.doi.org/10.1016/j. targeted to EGFR deliver antitumor microRNA to breast cancer cells, Mol. Ther. 21
nano.2015.10.012 [Epub ahead of print]. (2013) 185191.
[49] J. Wahlgren, L.K.T. De, M. Brisslert, F. Vaziri Sani, E. Telemo, P. Sunnerhagen, H. [73] K.J. Widder, A.E. Senyei, D.F. Ranney, Magnetically responsive microspheres and
Valadi, Plasma exosomes can deliver exogenous short interfering RNA to monocytes other carriers for the biophysical targeting of antitumor agents, Adv. Pharmacol.
and lymphocytes, Nucleic Acids Res. 40 (2012) e130. Chemother. 16 (1979) 213271.
[50] S.A. Kooijmans, S. Stremersch, K. Braeckmans, S.C. de Smedt, A. Hendrix, M.J. Wood, [74] A.E. Senyei, S.D. Reich, C. Gonczy, K.J. Widder, In vivo kinetics of magnetically
R.M. Schiffelers, K. Raemdonck, P. Vader, Electroporation-induced siRNA precipita- targeted low-dose doxorubicin, J. Pharm. Sci. 70 (1981) 389391.
tion obscures the efciency of siRNA loading into extracellular vesicles, J. Control. [75] A.K. Silva, N. Luciani, F. Gazeau, K. Aubertin, S. Bonneau, C. Chauvierre, D. Letourneur,
Release 172 (2013) 229238. C. Wilhelm, Combining magnetic nanoparticles with cell derived microvesicles for
[51] N.G. Stoicheva, S.W. Hui, Electrofusion of cell-size liposomes, Biochim. Biophys. Acta drug loading and targeting, Nanomed.: Nanotechnol., Biol. Med. 11 (2015) 645655.
1195 (1994) 3138. [76] H. Maeda, Macromolecular therapeutics in cancer treatment: the EPR effect and be-
[52] J.L. Hood, M.J. Scott, S.A. Wickline, Maximizing exosome colloidal stability following yond, J. Control. Release 164 (2012) 138144.
electroporation, Anal. Biochem. 448 (2014) 4149. [77] A. Mizrak, M.F. Bolukbasi, G.B. Ozdener, G.J. Brenner, S. Madlener, E.P. Erkan, T. Strobel,
[53] M.J. Haney, N.L. Klyachko, Y. Zhao, R. Gupta, E.G. Plotnikova, Z. He, T. Patel, A. X.O. Breakeeld, O. Saydam, Genetically engineered microvesicles carrying suicide
Piroyan, M. Sokolsky, A.V. Kabanov, E.V. Batrakova, Exosomes as drug delivery vehi- mRNA/protein inhibit schwannoma tumor growth, Mol. Ther. 21 (2013) 101108.
cles for Parkinson's disease therapy, J. Control. Release 207 (2015) 1830. [78] Y. Zhang, L. Li, J. Yu, D. Zhu, X. Li, H. Gu, C.Y. Zhang, K. Zen, Microvesicle-mediated
[54] G. Fuhrmann, A. Serio, M. Mazo, R. Nair, M.M. Stevens, Active loading into extracel- delivery of transforming growth factor beta1 siRNA for the suppression of tumor
lular vesicles signicantly improves the cellular uptake and photodynamic effect of growth in mice, Biomaterials 35 (2014) 43904400.
porphyrins, J. Control. Release 205 (2015) 3544. [79] S.C. Jang, O.Y. Kim, C.M. Yoon, D.S. Choi, T.Y. Roh, J. Park, J. Nilsson, J. Lotvall, Y.K. Kim,
[55] O.P. Wiklander, J.Z. Nordin, A. O'Loughlin, Y. Gustafsson, G. Corso, I. Mager, P. Vader, Y.S. Gho, Bioinspired exosome-mimetic nanovesicles for targeted delivery of che-
Y. Lee, H. Sork, Y. Seow, N. Heldring, L. Alvarez-Erviti, C.E. Smith, K. Le Blanc, P. motherapeutics to malignant tumors, ACS Nano 7 (2013) 76987710.
Macchiarini, P. Jungebluth, M.J. Wood, S.E. Andaloussi, Extracellular vesicle in vivo [80] J. Yoon, W. Jo, D. Jeong, J. Kim, H. Jeong, J. Park, Generation of nanovesicles with
biodistribution is determined by cell source, route of administration and targeting, sliced cellular membrane fragments for exogenous material delivery, Biomaterials
J. Extracellular Vesicles 4 (2015) 26316. 59 (2015) 1220.

You might also like