functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7jz9r5yfb.pdf ·...

47
Accepted Manuscript Functionalized extracellular vesicles as advanced therapeutic nanodelivery systems Mei Lu, Haonan Xing, Zhe Xun, Tianzhi Yang, Xiaoyun Zhao, Cuifang Cai, Dongkai Wang, Pingtian Ding PII: S0928-0987(18)30214-8 DOI: doi:10.1016/j.ejps.2018.05.001 Reference: PHASCI 4512 To appear in: European Journal of Pharmaceutical Sciences Received date: 27 March 2018 Revised date: 1 May 2018 Accepted date: 3 May 2018 Please cite this article as: Mei Lu, Haonan Xing, Zhe Xun, Tianzhi Yang, Xiaoyun Zhao, Cuifang Cai, Dongkai Wang, Pingtian Ding , Functionalized extracellular vesicles as advanced therapeutic nanodelivery systems. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Phasci(2017), doi:10.1016/j.ejps.2018.05.001 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Upload: others

Post on 19-Oct-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

Accepted Manuscript

Functionalized extracellular vesicles as advanced therapeuticnanodelivery systems

Mei Lu, Haonan Xing, Zhe Xun, Tianzhi Yang, Xiaoyun Zhao,Cuifang Cai, Dongkai Wang, Pingtian Ding

PII: S0928-0987(18)30214-8DOI: doi:10.1016/j.ejps.2018.05.001Reference: PHASCI 4512

To appear in: European Journal of Pharmaceutical Sciences

Received date: 27 March 2018Revised date: 1 May 2018Accepted date: 3 May 2018

Please cite this article as: Mei Lu, Haonan Xing, Zhe Xun, Tianzhi Yang, Xiaoyun Zhao,Cuifang Cai, Dongkai Wang, Pingtian Ding , Functionalized extracellular vesicles asadvanced therapeutic nanodelivery systems. The address for the corresponding authorwas captured as affiliation for all authors. Please check if appropriate. Phasci(2017),doi:10.1016/j.ejps.2018.05.001

This is a PDF file of an unedited manuscript that has been accepted for publication. Asa service to our customers we are providing this early version of the manuscript. Themanuscript will undergo copyediting, typesetting, and review of the resulting proof beforeit is published in its final form. Please note that during the production process errors maybe discovered which could affect the content, and all legal disclaimers that apply to thejournal pertain.

Page 2: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

1

Functionalized extracellular vesicles as advanced therapeutic nanodelivery systems

Mei Lu a, Haonan Xing a, Zhe Xun b, Tianzhi Yang c, Xiaoyun Zhao d, Cuifang Cai a, Dongkai

Wang a,*, Pingtian Ding a,**

a School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China

b China Institute of Metabolic Disease Research and Drug Development, China Medical

University, Shenyang, China

c Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University,

Bangor, ME, USA

d School of life Science and Biopharmaceutics, Shenyang Pharmaceutical University,

Shenyang, China

* Address for correspondence:

Dongkai Wang, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua

Road, Shenyang 110016, China. Tel: +86 24 23986310; +86 13998210961 (mobile). Fax: +86 24

23986310. E-mail: [email protected]. (D. Wang).

Pingtian Ding, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua

Road, Shenyang 110016, China. Tel: +86 24 23986305; +86 13940375008 (mobile). Fax: +86 24

23986305. E-mail: [email protected]. (P. Ding).

ACCEPTED MANUSCRIPT

Page 3: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

2

Abstract

Extracellular vesicles (EVs) are membrane enclosed vesicles that are shed by almost all cell

types, and play a fundamental role in cell-to-cell communication. The discovery that EVs are

capable of functionally transporting nucleic acid- and protein- based cargoes between cells,

rapidly promotes the idea of employing them as drug delivery systems. These endogenous vesicles

indeed hold tremendous promise for therapeutic delivery. However, issues associated with

exogenously administered EVs, including rapid clearance by the immune system, apparent lack of

targeting cell specificity, and insufficient cytoplasmic delivery efficiency, may limit their

therapeutic applicability. In this review, we discuss recent research avenues in EV-based

therapeutic nanodelivery systems. Furthermore, we narrow our focus on the development of

modification strategies to enhance the delivery properties of EVs, and elaborate on how to

rationally harness these functionalized vesicles for therapeutic delivery.

Keywords: Functionalization; Extracellular vesicles; Therapeutic nanodelivery systems; Targeting

capacity; Cytoplasmic delivery

ACCEPTED MANUSCRIPT

Page 4: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

3

Abbreviations: AA-PEG, aminoethylanisamide-polyethylene glycol; AD, Alzheimer's disease;

BBB, blood-brain barrier; BDNF, brain derived neurotrophic factor; CML, Chronic Myeloid

Leukemia; CPPs, cell penetrating peptides; c(RGDyK, cyclo(Arg-Gly-Asp-D-Tyr-Lys) peptide;

DSPE, 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine; EGFR, endothelial growth factor

receptor; EVs, Extracellular vesicles; FA, folate; GALA, GALA peptide; GPI,

glycosylphosphatidylinositol; ICAM-1, intercellular adhesion molecule 1; imDCs, immature

dendritic cells; IL3, Interleukin 3; iRGD, internalizing arginine-glycine-asparagine; ISEV,

International Society for Extracellular Vesicles; Lamp2b, lysosome-associated membrane

glycoprotein 2b; LFA-1, lymphocyte function-associated antigen 1; MPS, mononuclear phagocyte

system; MSCs, mesenchymal stromal stem cells; MVBs, multivesicular bodies; PDGFR,

platelet-derived growth factor receptor; PEG, polyethylene glycol; PPI, proton pump inhibitors;

RVG, rabies viral glycoprotein; SIRPα, signal regulatory protein α; SMCNCs, superparamagnetic

magnetite colloidal nanocrystal clusters; VEGF, vascular endothelial growth factor; VSV-G, G

protein of vascular stomatitis virus; WFA, Withaferin A.

ACCEPTED MANUSCRIPT

Page 5: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

4

1. Introduction

Extracellular vesicles (EVs), also termed as natural liposomes, are small membrane-enclosed

vesicles shed in an evolutionally conserved manner by cells ranging from prokaryotes to higher

eukaryotes (Colombo et al., 2014). With a size ranging from 40 to 1000 nm, EVs are composed of

a lipid bilayer decorated with various transmembrane proteins (Raposo and Stoorvogel, 2013;

Tkach and Thery, 2016). As defined primarily by subcellular origin, cells can secrete a multiple

subset of EVs. Among which, the most interest is focused on two major groups of EVs: exosomes

and microvesicles. Exosomes are originated from intraluminal budding of multivesicular bodies

(MVBs), while microvesicles are formed by directly budding from the plasma membrane (Thery

et al., 2009; Thery et al., 2002). EVs are capable of transferring biologically active molecules

between cells locally and at distance, thereby regulating gene expression and cellular function in

recipient cells (Valadi et al., 2007; van Dommelen et al., 2012) As such, these vesicles are

endorsed with a vast array of functions in a multitude of physiological processes and pathologies

(Baj-Krzyworzeka et al., 2007; Yanez-Mo et al., 2015), which has triggered new developments in

the field of diagnostics. Actually, the use of EVs as disease biomarkers has become an area of

intense investigation, given that healthy subjects and patients may secrete EVs with different

contents (Andreu et al., 2017; Barile and Vassalli, 2017). Moreover, giving the increased release of

EVs by diseased cells (cancer cells), recent clinical reports proposed to use the level of EVs

released into human body fluids as a reliable biomarker of diseases, specifically of cancers

(Cappello et al., 2017; Fais et al., 2016). For example, melanoma patients and prostate cancer

patients have remarkably higher levels of EVs in the plasma than healthy subjects, it is promising

to quantify EVs in the plasma of these patients and harness these endpoints in early diagnosis

(Logozzi et al., 2017; Logozzi et al., 2009). In addition to serving as disease biomarkers, EVs are

also emerging as a very valuable class of drug nanocarriers. In the last few years, excitement was

created to harness EVs as therapeutic nanodelivery systems because of their natural role in

transporting biological molecules amongst cells (Fuhrmann et al., 2015; Jiang et al., 2017). EVs

have been extensively studied as biological nanocarriers for a wide array of therapeutics, ranging

from small chemotherapeutic molecule to macromolecular miRNA, siRNA, DNA, and proteins in

various preclinical studies (Srivastava et al., 2016; Sun et al., 2010).

The therapeutic use of EVs as nanocarriers is boosted by their low immunogenicity and

ACCEPTED MANUSCRIPT

Page 6: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

5

toxicity but high biological permeability to cross the blood-brain barrier (BBB) and

biocompatibility (El Andaloussi et al., 2013). Despite of these remarkable features, major

challenges still remain to be addressed before EVs can be used as competitive therapeutic

nanocarriers. In addition to issues concerning lack of efficient methods for isolation, loading,

scalable production, and quality regulation of EVs (Fais et al., 2016; Lener et al., 2015; Lu et al.,

2017), the intracellular delivery efficiency of EVs still remains unsatisfactory, especially after

systemic administration (Mulcahy et al., 2014; Nakase and Futaki, 2015; Smyth et al., 2015). For

efficient systemic drug delivery, therapeutic nanocarriers should possess three crucial features:

sufficiently long circulation time for effective tissue/organ accumulation, satisfactory penetration

and homing capacity to reach target tissues, and high cellular uptake and efficient endosomal

escape to achieve efficient cytoplasmic delivery (Haussecker, 2014). However, the unfavorable

pharmacokinetic profile, lack of targeting cell selectivity, and insufficient cytosolic delivery

efficiency of unmodified EVs may prohibit them from becoming clinically acceptable therapeutics.

For instance, naturally secreted EVs with no modifications to their composition may not readily

avoid the mononuclear phagocyte system (MPS) sequestration as assumed from their “self” nature

(van der Meel et al., 2014). After systemic administration, unmodified EVs have been described to

undergo swift removal from the circulation mediated by the MPS, which substantially limit their

accumulation in tissues of interest (Takahashi et al., 2013). In addition, EVs released by most cells

exhibit limited tropism to a specific cell type due to their highly complex and variable

composition (Barile and Vassalli, 2017). This may result in off-target effects when EVs are applied

for therapeutic delivery. Furthermore, the efficiency of these vesicles to overcome membrane

barriers, such as crossing the plasma membrane, and endosomal escape for cytoplasmic release of

their payloads, is still considered insufficient for therapeutic application . There is a competition

for the cellular uptake of these vesicles, as a considerable number of endogenous EVs are secreted

into the bodily fluids. On the other hand, the negative charge of EV membrane may also prevent

them from binding to negatively charged plasma membranes. Additionally, after internalization by

the recipient cell, EVs require endosomal escape from degradation by lysosomes, however, their

cytoplasmic release efficiency remains insufficient (Nakase and Futaki, 2015; Nakase et al., 2017).

To this end, strategies for enhancing the delivery properties of systemically delivered EVs are

greatly needed.

ACCEPTED MANUSCRIPT

Page 7: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

6

Taking advantage of the possibility to modulate EV composition, the potential of EVs as drug

delivery vehicles can be further improved by incorporation of functional moieties. Recently,

strategies to modify the surface of EVs or encapsulate them with exogenous materials for better

delivery properties have been widely reported (Koh et al., 2017; Nakase and Futaki, 2015; Silva et

al., 2015; Tian et al., 2014). For example, it is plausible to decorate EVs with “stealth” polymer

coatings, such as polyethylene glycol (PEG) and dextran sulfate, typically applied in synthetic

nanoparticles, to provide them with greater disguise from MPS and extend their circulation

half-life (Kooijmans et al., 2016a; Watson et al., 2016). Additionally, engineering EVs to express

peptides, such as rabies viral glycoprotein (RVG) that binds to acetylcholine receptor expressed on

neuronal cells, enables EVs to efficiently pass through the blood-brain barrier and target siRNA to

the brain (Alvarez-Erviti et al., 2011). As another example, conjugating EVs with arginine-rich

cell penetrating peptides (CPPs) could activate the macropinocytosis pathway for effective cellular

uptake, thus enhancing the cytosolic delivery efficiency (Nakase et al., 2016). Generally,

incorporation of functional moieties on EV surface can be accomplished by using “cell

engineering” techniques, wherein donor cells are transfected with recombinant plasmids encoding

EVs’ membrane proteins (e.g. Lamp2b) fused to specific functional molecules. Alternatively,

functionalized EVs may be generated by employing “EV engineering”, wherein EVs are directly

modified through mechanisms of biochemical conjugation, post insertion, or electronic interaction.

In this review, we provide an update overview of EV-based therapeutic nanodelivery platforms

with focus on EV modification strategies, in order to efficiently harness functionalized EVs for

therapeutic delivery.

2. Unmodified EVs for therapeutic delivery

The advent of nanotechnology has brought a rapidly growing body of new nanoscaled drug

delivery vehicles, such as the most investigated lipid- and polymer-based nanocarriers (Duncan,

2003; Puri et al., 2009). Both of these nanoparticles have been effectively used to deliver a wide

range of therapeutics. However, they often fail to merge drug delivery efficiency with

biocompatibility, thus confronting issues such as potential toxicity and immunogenicity (Kumari

et al., 2010; Raemdonck et al., 2014). In this circumstance, EVs are increasingly emerged as a

fascinating drug delivery vehicle that outperforms synthetic nanoparticles. EVs have been shown

with strong protection for their contents and high permeability to traverse biological barriers, such

ACCEPTED MANUSCRIPT

Page 8: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

7

as the BBB (Yang et al., 2017b). More importantly, These vesicles have been proposed to be less

immunogenic and toxic but with higher biodegradability than artificial delivery vehicles, possibly

due to their biological origin (Johnsen et al., 2014; Lai et al., 2013). These properties facilitate the

development of EVs as suitable nanocarriers for therapeutics and potential avenues for the

treatment of a number of diseases.

Many delivery approaches that take advantage of the innate delivery properties of EVs have

shown increased therapeutic efficacy (Martins-Marques et al., 2016; Momen-Heravi et al., 2014;

Sterzenbach et al., 2017). For instance, doxorubicin was successfully loaded into breast cancer

cell-derived EVs, which was more efficient than free drug in the treatment of breast cancer and

ovarian cancer mouse model. EVs loaded with doxorubicin displayed increased stability and

accumulation in tumor tissue, leading to a significant suppression of tumor growth and reduced

cardiotoxicity in mouse, a main side-effect of doxorubicin (Hadla et al., 2016). In a recent study,

Yang et al. demonstrated EVs from brain endothelial cells can ferry doxorubicin and paclitaxel

across the BBB in a zebra fish model, leading to the inhibition of tumor progression (Yang et al.,

2015). Whereas, both free drugs remained localized in the vasculature and could not penetrate the

BBB. Interestingly, bovine milk-derived EVs can also function as efficient carriers for

chemotherapeutics. Munagala et al. showed that EVs loaded with Withaferin A (WFA), not only

exhibited significantly higher efficacy in in vitro cell culture, but also possessed greater tumor

inhibitory effect in in vivo tumor models compared with free WFA (46% vs 23%) (Munagala et al.,

2016). Additionally, EVs possess the ability to deliver photodynamic molecules, thus holding

great potential in photodynamic therapy for cancer treatment (Kusuzaki et al., 2017). For instance,

Lessi et al. recently showed that EVs loaded with acridine orange exhibited increased

effectiveness and decreased toxicity in human melanoma cells, as compared to free acridine

orange. This was a completely new prototype for harnessing EVs in the treatment of cancers (Iessi

et al., 2017). These results confirmed EV formulations could be effectively used to enhance the

therapeutic efficacy of small chemotherapeutic molecules. However, it is noteworthy that

extracellular acidity probably represents the most important phenotype of malignant tumors (Fais

et al., 2014; Spugnini and Fais, 2017). This is a key point to tailor exosomes for anticancer therapy,

because it has been widely shown that the acidic milieu of cancer may serve as a selective

pressure to trigger the release of highly level of EVs by cancer cells, leading to cancer progression

ACCEPTED MANUSCRIPT

Page 9: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

8

and metastasis (Logozzi et al., 2017; Parolini et al., 2009). To this end, it appears that antiacidic

molecules such as proton pump inhibitors (PPI) and buffers such as sodium bicarbonate and citrate,

may be utilized to improve the efficacy of anticancer therapy (Fais et al., 2014; Spugnini and Fais,

2017). For example, Federici and coworkers showed that PPI pretreatment can increase the

cellular uptake of cisplatin compared with untreated cells, leading to a significant inhibition of EV

release and a clear suppression of tumor growth both in vitro and in vivo (Federici et al., 2014).

Other than chemotherapeutics, unmodified EVs have also been extensively exploited to

shuttle small RNAs that have poor stability in the circulation. Yang’s group further employed brain

endothelial EVs to deliver vascular endothelial growth factor (VEGF) siRNA across the BBB to

the brain of zebra fish. Both in vitro and in vivo results showed siRNA was efficiently transported

to the brain and inhibited the tumor growth (Yang et al., 2017b). Similarly, marrow MSC-derived

EVs were therapeutically used to deliver miRNA (miR-146) to induce regression of glioma

growth (Katakowski et al., 2013). Despite of these encouraging results, Stremersch and coworkers

demonstrated that EVs appeared to be less effective than conventional liposome formulations in

the functional delivery of small RNAs. They reported evidence that EVs were unable to

functionally deliver cholesterol-conjugated siRNA and endogenous miRNA to mediate target gene

knockdown despite robust cellular uptake. In contrast, conventional anionic fusogenic liposomes

were capable of inducing a marked gene knockdown under equal experimental conditions. The

authors proposed that endolysosomal entrapment might be a main barrier that resulted in the

incapacity of unmodified EVs to guide cytosolic delivery of small RNAs (Stremersch et al., 2016).

While EVs have been widely explored to deliver small molecular drugs and RNAs, there are some

evidences that EV-mediated transfer of macromolecular DNA and protein is also possible. For

instance, Lamichhane and coworkers showed that DNA loading into EVs by electroporation

seemed to be size-dependent. Linear DNA above 750bp and plasmid DNA above 4.5kb was

observed to have very low loading efficiency (Lamichhane et al., 2015). Similarly, successful

loading of DNA and feasible cellular uptake of drug loaded EVs did not lead to transcription and

functional gene expression, underscoring other factors may limit the cytosolic delivery of EVs’

cargoes. Intracellular delivery of bioactive proteins is an attractive tool to replace missing or

poorly expressed proteins. In recent years, there is a growing passionate investigation bustle to

enrich proteins in EVs for improved delivery efficiency. For instance, Yuan et al. demonstrated

ACCEPTED MANUSCRIPT

Page 10: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

9

that EVs derived from macrophages expressed the integrin lymphocyte function-associated

antigen 1 (LFA-1) and intercellular adhesion molecule 1 (ICAM-1), could deliver a cargo protein,

the brain derived neurotrophic factor (BDNF), traversing the BBB of mice after intravenous

administration. Moreover, an observable enhancement of delivery occurred during brain

inflammation, a common condition associated with many brain diseases (Yuan et al., 2017).

Collectively, EVs are characterized by several favorable features that highlight eloquently in

most studies where EVs may serve as powerful therapeutic nanocarriers. However, in some

circumstances they exhibited low capability of delivering their cargoes. To date, it still remains

elusive to explain why under some conditions EVs were very efficient for drug delivery, while in

other circumstance they exhibited dysfunctional. To this end, future research should be dedicated

towards elucidating the cellular mechanism behind successful EV-mediated delivery. Yet some

obstacles may lay behind these divergent results that need to be overcome for translation of EVs

into clinical therapies. Currently, no standard techniques have been established for the clinical

grade production, storage, and quality control of EV-based therapeutics (Fais et al., 2016; Lener et

al., 2015). In addition, safety issues, including general toxicity, potential immunogenicity,

immunotoxicity, and tumourigenicity, must be highlighted for therapeutic application of EVs in a

clinical setting (Lener et al., 2015). However, clinical translation of EVs is a long-standing issue,

which requires cooperation between researchers, clinicians, and competent authorities in EV

research field. Luckily, as a consequence of the substantial advance in this field, a level of

consensus on these challenges have been achieved by the International Society for Extracellular

Vesicles (ISEV), although it has not yet been fully implemented in clinical studies (Fais et al.,

2016). In addition to these considerable challenges that must be highlighted, strategies to

overcome several preclinical challenges of EVs, including short circulation time, apparent lack of

specific cell tropism, and insufficient cytoplasmic delivery efficiency are also essential to facilitate

the clinical translation of EVs (Alvarez-Erviti et al., 2011; Nakase et al., 2016; Smyth et al., 2015).

To this end, improving the delivery properties of EVs via surface modification or encapsulation

with exogenous materials is awaited with great interest to deal with these sticky issues, as they are

warranted to increase the circulation stability of intravenously injected EVs, improve their

targeting capability for better reaching tissues of interest, and facilitate EV payload delivery to the

cytosol of target cells. Rather encouragingly, the possibility to modify these vesicles and

ACCEPTED MANUSCRIPT

Page 11: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

10

functionalize them with various functional moieties offers exiting opportunities to extend the

delivery potential of EVs beyond their native capability.

3. Modification techniques to enhance EVs’ delivery properties

For therapeutic applications, EVs need to be functionalized with specific moieties to improve

their delivery properties. In the case of synthetic nanocarriers, this is generally accomplished

through direct introduction of functional ligands on the particle surface. However,

functionalization appears to be more complex for EVs as their composition is dictated by the

donor cells. Nevertheless, multiple modification strategies have been successfully applied to

enhance the intracellular delivery efficiency of EVs (Kooijmans et al., 2016a; Tamura et al., 2017;

Tian et al., 2014). Generally, EV modification can be classified into “cell engineering” and “EV

engineering”. Provided that molecules delivered to cell membrane are naturally incorporated in

the budding microvesicles, while materials internalized inside the cells can be packaged into the

secreted exosomes, it can take advantage of such a biosynthesis process for EV modification by

genetic manipulation of their parent cells. Such a modification approach is called “cell

engineering”. While in “EV engineering”, surface functionalization is accomplished by direct

manipulation of EVs (Fig. 1). In this section, we will discuss these two EV modification

techniques.

3.1. Genetic engineering of producing cells for EV modification

“Cell engineering” hijacks a cellular biosynthesis process to generate modified EVs. By

using the cell’s own machinery for protein production, this approach allows for the expression of

proteins or peptides on EV surface with preserved direction and function (Zhao et al., 2016). A

general strategy of “cell engineering” is to fuse ligands with specific function to EV-enriched

membrane proteins. For example, in an early proof-of-concept study, neuron-targeting EVs were

generated by fusing RVG peptide with lysosome-associated membrane glycoprotein 2b (Lamp2b),

an EVs-associated membrane protein (Alvarez-Erviti et al., 2011). Likewise, engineering

immature dendritic cells (imDCs) to express a fusion construct of Lamp2b and

αν-integrin-specific iRGD peptide has been exploited to produce iRGD modified EVs (Tian et al.,

2014). However, lamp2b display of a muscle-specific peptide was shown to be minimally

effective at targeting muscle cells, illustrating a variable performance of this approach

(Alvarez-Erviti et al., 2011). Furthermore, concerns have also been raised about the long stability

ACCEPTED MANUSCRIPT

Page 12: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

11

of Lamp2b-fused ligands, as the fused peptides tend to be proteolytic degradation in the endosome

before being sorted into EVs (Hung and Leonard, 2015). To suppress peptide loss, Hung and

coworkers pointed out that the introduction of a glycosylation motif on Lamp2b can protect

targeting construct from degradation, hence improving EV delivery to neuroblastoma cells in vivo

(Hung and Leonard, 2015).

In addition to Lamp2b, other EV-associated membrane proteins, such as platelet-derived

growth factor receptor (PDGFR), lactadherin, and Cytolysin A, have also been used as anchors

and fusion partners for protein domains/peptides of interest. Fusing functional ligands to the

transmembrane domain of PDGFR is a fascinating display system for EV modification (Ohno et

al., 2013). Through this method, EVs expressing endothelial growth factor receptor

(EGFR)-targeting peptide anchored by the transmembrane receptor of PDGF showed significantly

improved cell association to EGFR-expressing tumor cells (Ohno et al., 2013). In addition, the

C1C2 domain of lactadherin is another commonly used anchor for EV display of functional ligand

(e.g. antibodies against tumor biomarkers) (Delcayre et al., 2005). Recently, a similar approach

has been used to equip EVs with enhanced targeting properties. In this method, Escherichia coli

was engineered to express fusions between anti-HER2 affibody and the C-terminus of Cytolysin A,

which led to display of anti-HER2 affibody on the secreted EVs (Gujrati et al., 2014). However,

fusion of recombinant proteins or functional peptides to EV-specific membrane proteins may

impair the structural and functional integrity of these membrane proteins (Kooijmans et al., 2016c).

In this circumstance, a new EV marker glycosylphosphatidylinositol (GPI) can function as an

alternative anchor for the display of functional ligands. With such an approach, functional ligands

can be localized on GPI-rich EV surface through engineering EV-producing cells to express EGFR

nanobodies fused to GPI-anchor, to target cancer cells overexpressing higher level of EGFR

(Kooijmans et al., 2016a). Given that EVs are characterized to contain tightly packed membrane

domains (lipid rafts) which are enriched in GPI, GPI-fused nanobodies were strongly enriched in

EVs compared with their parent cells (Haraszti et al., 2016). Moreover, GPI anchoring may

potentially function as a versatile tool to incorporate a wide range of functional ligands, such as

antibodies, reporter proteins, and immune stimulatory molecules (Kooijmans et al., 2016a).

To address the limitation of conventional “cell engineering” methods, new EV

functionalization strategies have been explored. A novel approach is to produce fusogenic

ACCEPTED MANUSCRIPT

Page 13: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

12

liposomes containing desired ligands, deliver them to cells through membrane fusion, and then

EVs equipped with functional ligands could be harvested (Lee et al., 2016). This method enables

an efficient and controlled manner to arm EVs with various functional ligands, such as peptides

and bio-orthogonal chemicals, without modification of EVs’ membrane proteins. In addition, such

a liposome-based cellular engineering method allows simultaneous modification of EVs with

multiple ligands with disparate functions (Lee et al., 2016). Additionally, surface modification can

also be achieved by simple incubating cells with, mainly hydrophobic, ligand of interest, and then

ligand may interact with the cell phospholipid bilayers driving spontaneously membrane insertion.

For example, lipidated AS1411 aptamer could spontaneously insert into cellular lipid bilayer,

resulting in efficient immobilization of targeting ligands onto cell membrane by simple incubation

(Wan et al., 2018). Afterwards, aptamer-functionalized EVs could be generated by repeated

mechanical extrusion of cells through multiple filters with different pore size. Overall, mechanical

extrusion approach may represent a rapid, easy, and economic technique to produce functionalized

EVs (Wan et al., 2018). Moreover, a completely different approach to surface engineering is to

encapsulate exogenous materials into cells, which can then be packaged into the secreted EVs. For

example, magnetically and optically targeting responsive EVs can be generated by incubating

macrophages with iron oxide nanoparticles and small molecule photosensitizers (Silva et al., 2013;

Silva et al., 2015). Then modified EVs could target specific areas of the body to achieve magnetic

and photodynamic targeting by applying an external magnetic field and optical stimulation.

However, challenges of this approach include difficulties in targeting deep tissues in the body, and

possible toxicity concerns associated with the administration of magnetic nanoparticles and

photosensitizers (Clavijo-Jordan et al., 2012).

3.2. Direct engineering of EVs

Conventional cell-based EV functionalization strategies are frequently criticized for

inefficiency, as only a low fraction of functional materials can be displayed on EV surface (Kim

and Eberwine, 2010). In addition, their production protocols are reported to be complex,

time-consuming, and not be readily applicable to all ligands, such as toxic proteins (Armstrong et

al., 2017). In contrast, direct EV engineering offers a more controllable and efficient tool to

incorporate high densities of functional moieties regardless of ligand type (Armstrong et al., 2017).

In addition, EVs are nonliving entities, hence, it is possible to use some chemical reagents and

ACCEPTED MANUSCRIPT

Page 14: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

13

reaction conditions that could not be used in “cell engineering”. Nevertheless, there are still some

constraints, for example, the reaction conditions must be well controlled to avoid exposing EVs to

excessive solvent, temperature or pressure, as which may impair membrane integrity and denature

the functionality of membrane proteins (Wang et al., 2014).

Direct “EV engineering” is typically achieved by using covalent bioconjugation or

noncovalent modification, such as post insertion and electronic interaction. In covalent

conjugation strategies, chemical reactions are directly performed at EV surface under

biocompatible reaction conditions (Smyth et al., 2014). By employing chemical linkers, functional

ligands can be linked to amines, a reactive functional group expressed on EV membrane. This

reaction mechanism has been used to graft alkyne moieties onto EV surface. After conjugation

with membrane amines, the introduction of reactive alkyne bases allows a second click chemistry

reaction with model azide compounds (e.g. Azide-Fluor 545). As a consequence, functional

ligands can be stably conjugated onto the surface of isolated EVs (Smyth et al., 2014). As a

proof-of-principle, the result demonstrated chemical reactions were well suited for EV

modification, without significant effects on the structure integrity of EVs or their interaction with

recipient cells. Similarly, EVs could also be conjugated with cyclo(Arg-Gly-Asp-D-Tyr-Lys)

peptides [c(RGDyK)], a ligand with high affinity to integrin αvβ3 by using click chemistry

(bioorthogonal copper-free azide alkyne cyclo-addition). This technique was demonstrated to be

useful for rapid and large-scale production of functionalized EVs (Tian et al., 2018). In contrast to

covalent modification strategies, noncovalent functionalization approaches involve relatively mild

reaction conditions. As for post insertion, hydrophobic or lipids-conjugated ligands could

spontaneously insert into EV lipid bilayer through hydrophobic interaction. Through this approach,

nanobodies specific for EGFR were conjugated to the distal ends of lipid-PEG derivatives, which

formed nanobody-PEG-micelles in aqueous solution. Then, the resulted micelles were mixed with

EVs and incubated at elevated temperatures to facilitate micelle disintegration and spontaneous

integration of nanobody-PEG-lipids onto EV membrane. As a consequence, a

temperature-dependent transfer of nanobody-PEG-lipids to EV membrane was observed,

suggesting a post-insertion mechanism (Kooijmans et al., 2016b). It is noteworthy that post

insertion possesses general applicability, given that EVs from Neuro2A cells and platelets shared

similar transfer pattern of nanobody-PEG-lipids. In addition, the ease of ligand conjugation to

ACCEPTED MANUSCRIPT

Page 15: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

14

PEGylated phospholipids makes it applicable to a wide range of functional ligands, such as

antibodies and peptides (Kooijmans et al., 2016b). Another noncovalent modification method is

based on electrostatic interaction. EVs are negatively charged with a zeta potential of

approximately -11.90mV (Saari et al., 2015), which can be exploited to equip EVs with cationic

moieties. For example, EV modification was achieved by simple mixing purified EVs with

cationized pullulan through an electrostatic interaction between both substances. In this approach,

pullulan was cationized with spermine to enhance its electrostatic interaction with the negatively

charged surface of EVs (Tamura et al., 2017). The advantages and disadvantages of “cell

engineering” and “EV engineering” are shown in Table 1.

4. Modified EVs for therapeutic delivery

Provided that unmodified EVs’ unfavorable characteristics may limit their widespread

application as drug delivery vehicles, EV modification could open up exciting opportunities for

improving their delivery capability and extending the repertoire of their payloads. The

introduction of function-specific moieties to EV surface might be an attractive strategy to endow

these vesicles with enhanced circulation time, increased targeting capacity, and improved

cytoplasmic delivery efficiency. Different strategies for EV functionalization are shown in Fig. 2.

In this section, we will elaborate on how to harness these modified EVs for efficient therapeutic

delivery.

4. 1. Modified EVs for extended circulation time

As EVs are abundantly present in our biological fluids, it seems logical to postulate that they

may be quite stable in the blood circulation when being used as therapeutic nanocarriers. However,

recent studies demonstrated that unmodified EVs from many cell lines all showed rapid clearance,

preferential accumulation in MPS associated organs, such as liver, spleen, and lungs, and followed

by swift elimination by bile excretion, renal filtration or phagocytosis by the immune system (Imai

et al., 2015; Saunderson et al., 2014; Smyth et al., 2015; Takahashi et al., 2013; Tian et al., 2014;

Wiklander et al., 2015). In the past decade, the fate of intravenously-injected EVs has been

extensively studied through different tools, such as luciferase labeling and fluorescence-mediated

imaging (Lai et al., 2014; Takahashi et al., 2013). Pharmacokinetic studies reported that EVs

abundance was decreased by more than a half from 30 to 60 minutes after systemic administration

(Lai et al., 2014). Recently, EVs were described to disappear very quickly from the circulation

ACCEPTED MANUSCRIPT

Page 16: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

15

with a half-life of approximately two minutes, and completely disappear from the blood stream at

four hours upon intravenous injection (Saunderson et al., 2014; Takahashi et al., 2013).

Additionally, biodistribution of unmodified EVs showed a rapid distribution phase in the spleen,

liver, lung, and kidney at approximately 30 minutes after systemic injection, and followed by a

longer distribution phase reflecting elimination through the liver and kidney in 1 to 6 hours upon

intravenous administration (Lai et al., 2015; Lai et al., 2014). Of note, EV clearance was observed

to be much slower in macrophage-depleted mice than that in groups not subjected to macrophage

depletion treatment, illustrating a macrophage-dependent clearance of intravenous-injected EVs

from the blood circulation (Imai et al., 2015). In addition, it is proposed that the immune

recognition of exogenously-administered EVs is partly mediated by the exposure of externalized

phosphatidylserine on EVs to macrophages (Deschout et al., 2014; Feng et al., 2010). Together,

these results indicate that EVs display an unfavorable pharmacokinetic profile resembles that of

synthetic nanoparticles. Systemically administered EVs may not readily avoid MPS sequestration

as speculated from their wide existence in the blood circulation and “self” nature.

Considering that the rapid clearance feature along with minimal tissue/organ accumulation of

unmodified EVs may limit their application as drug delivery vehicles. Strategies to protract the

circulation time of EVs are urgently required. A common used approach to prevent swift removal

of nanoparticles from circulation by opsonization is to decorate these particles in a PEG corona

(Caracciolo, 2015). PEGylation has been shown to be a powerful way to prolong the circulation

half life of drug-loaded liposomes (Perche and Torchilin, 2013). Recently, PEGylation has been

explored to increase the circulation time of EVs (Table 2). Kooijmans and coworkers showed that

insertion of PEG-lipids to EV membrane endowed them with stealth properties, and significantly

extended their circulation time in mice after intravenous injection (Fig. 3). To be specific,

nanobody-PEG EVs could be detectable in blood for longer than 60 minutes after intravenous

injection. Whereas, unmodified EVs were rapidly cleared from the circulation within 10 minutes.

Noteworthy, incorporation of PEG chain in EV membrane reduced their interactions with

non-targeted cells, while at the same time, display of nanobodies on vesicle surface enhanced EV

association with tumor cells expressing EGFR (Kooijmans et al., 2016b). In another study by Kim

et al., modification of EVs with aminoethylanisamide-polyethylene glycol vector moiety

(AA-PEG) was demonstrated to significantly increase EVs’ circulation time in the blood

ACCEPTED MANUSCRIPT

Page 17: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

16

circulation after systemic injection in mice, which allowed for robust accumulation in tumor and

greater in vivo therapeutic efficacy than non-modified EVs and free paclitaxel (Kim et al., 2018).

Based on these findings, modification of EVs with hydrophilic polymers though surface

engineering may prove crucial for prolonging EV circulation time, and represent a useful strategy

to the development of next generation of EV-based therapeutic nanodelivery systems. In addition,

concomitant functionalization of EVs with stealth properties and targeting capacity could enhance

EVs association with biomarkers of tumor cells, while at the same time reduce off-target effects.

However, it is of noted that PEG dilemma as observed in PEGylated liposomes, including reduced

cellular uptake/endosomal escape, and accelerated blood clearance after repeated administration,

could also occur to PEGylated EVs (Amoozgar and Yeo, 2012; Ishida et al., 2006). Therefore,

despite efficient shielding EVs from immune recognition through post insertion of PEG, it is

important to optimize several decoration parameters, such as grafting density and length of the

PEG chains. Another major consideration is that PEGylation greatly alters the surface composition

and behavior of EVs in vivo, which may lead to a question to what extent PEGylated EVs are

advantageous over synthetic nanocarriers. In this regard, future research should focus more on the

development of alternative functional ligands. For example, introduction of “self” proteins, such

as CD47, CD55, and CD59 to EV surface might help to protect them from phagocytotic clearance

and prolong circulation time (Clayton et al., 2003; Long and Beatty, 2013). In a recent study, EVs

were engineered to overexpress CD47, which interacted with SIRPα to produce a “don’t eat me”

signal in phagocytes, thus permitting EVs with a superior escape from phagocytosis by the MPS.

As a result, KRAS siRNA loaded EVs functionalized with CD47 suppressed multiple mouse

models of pancreatic cancer and significantly enhanced overall survival (Kamerkar et al., 2017).

4. 2. Modified EVs for improved targeting capability

Targeting delivery of drugs has been much sought to minimize off-target toxicity, reduce dose,

and enhance therapeutic efficacy. However, recent biodistribution studies revealed that after

systemic administration, naturally secreted EVs with no modifications accumulated predominantly

in MPS associated organs, such as the liver, spleen, and lung, and very few of EVs could be

delivered to target issues (Morishita et al., 2015; Ohno et al., 2013; Tian et al., 2014; Wiklander et

al., 2015). Such an unfavorable distribution characteristic may induce severe off-target effects and

limit the therapeutic application of unmodified EVs. Actually, EVs derived from most cells lack

ACCEPTED MANUSCRIPT

Page 18: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

17

efficient targeting of specific cell types, especially after systemic administration, and this may

have become one of the most challenging concern for EV-mediate drug delivery (Sullivan et al.,

2017). Therefore, the targeting properties of EVs need to be improved before generic adoption of

EVs as a competitive therapeutic delivery vehicle can be considered.

EVs may be endowed with improved targeting properties by displaying cell-specific targeting

ligands on their surface or packaging exogenous targeting materials. Some frequently used

homing ligands include peptides, such as RVG targeting acetylcholine receptors on neurons (Yang

et al., 2017a), αν-integrin-specific internalizing arginine-glycine-asparagine (iRGD) peptide

(Sugahara et al., 2009), GE11 peptide binding specifically to EGFR (Klutz et al., 2011), and

fragment of Interleukin 3 (IL3) targeting IL3 receptor (Nievergall et al., 2014), proteins like

nanobodies (Kooijmans et al., 2018), specific targeting molecules such as folate (FA) (Zhang et al.,

2017), and iron oxide (Qi et al., 2016) (Table 3). For peptide targeting, a widely used strategy is to

insert the coding sequence of a targeting peptide in-frame in-between the coding sequences of

signal peptides and EV associated membrane proteins (Alvarez-Erviti et al., 2011; Hartman et al.,

2011). An early demonstration was reported by Alvarez-Erviti and colleagues, who equipped EVs

with Lamp2b-RVG peptide through genetic engineering of the donor cells with plasmid construct.

RVG-modified EVs efficiently delivered siRNA across the BBB to target acetylcholine receptor in

the brain, and silenced a gene of relevance to Alzheimer's disease (AD) (Alvarez-Erviti et al.,

2011). Upon the first description of engineered EVs for enhanced targeting capacity, many studies

have assessed the potential of targeting delivery via peptide-modified EVs. RVG equipped EVs

were also applied to shuttle opioid receptor mu siRNA across the BBB targeting neurons in mouse

brain (Liu et al., 2015). Particularly, the targeting capacity of RVG-modified EVs was quantified

to be two-fold greater in regard to the accumulation in the brain than non-targeted EVs (Liu et al.,

2015). Through the same approach, targeting peptide iRGD was successfully introduced to EVs

derived from immature denderitic cells to target breast cancer (Tian et al., 2014). Ohno et al.

provided one of the earliest proof-of-concept studies, where EV producing cells were engineered

to express the transmembrane domain of PDGFR fused to the GE11 peptide, a synthetic peptide

that binds specifically to EGFR. GE11 modified EVs could efficiently deliver let-7a miRNA to

EGFR-expressing xenograft beast cancer tissue in mice, leading to a marked inhibition in tumor

growth (Ohno et al., 2013). Recently, EV producing cells were engineered to express Lamp2b

ACCEPTED MANUSCRIPT

Page 19: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

18

fused to a fragment of IL3. EVs equipped with IL3-Lamp2B loaded with Imatinib or with

BCR-ABL siRNA, were able to target Chronic Myeloid Leukemia (CML) cells and suppressed

cancer cell growth both in vitro and in vivo (Bellavia et al., 2017). More recently, Tian et al.

conjugated an integrin αvβ3-specific c(RGDyK) peptide onto EV surface via click chemistry. After

intravenous injection, c(RGDyK)-conjugated EVs could efficiently target the lesion region of

ischemic brain in mice, resulting in a significant suppression of the inflammatory response and

cellular apoptosis in the lesion region (Tian et al., 2018). Taken together, there are high

expectations that further development of peptide-modified EVs may help to usher in efficient

EV-based targeting delivery paradigms.

Apart from peptides, incorporating targeting proteins, such as nanobodies and SIRPα, to EV

surface has also been demonstrated as an attractive targeting strategy (Bryniarski et al., 2013; Koh

et al., 2017; Kooijmans et al., 2016a). Recently, Kooijmans et al. transfected EV producing cells

with vectors encoding EGFR nanobodies fused to GPI anchor signal peptides to target

EGFR-overexpressing tumor cells. Display of GPI-linked EGFR nanobodies on vesicle surface

significantly improved EV binding to tumor cells, which was dependent on EGFR density

(Kooijmans et al., 2016a). Alternatively, EGFR nanobody conjugated to PEG was introduced to

EV membrane via post-insertion mechanism, which greatly enhanced EVs binding to EGFR-

overexpressing tumor cells (Kooijmans et al., 2016b). More recently, Koh et al. generated a rather

sophisticated fusion protein containing SIRPα on top of the PDGFR. By applying PDGFR as a

pedestal, SIRPα was expressed on the surface of resulting EVs. SIRPα functionalized EVs

interfered CD47-SIRPa interaction between CD47-overexpressing cancer cells and bone marrow

derived macrophages, leading to enhanced tumor phagocytosis and significant inhibition of tumor

growth. The result manifested that modified EVs targeting to CD47-overexpressing cancer cells

may represent a versatile tool for cancer immunotherapy (Koh et al., 2017).

Additionally, EV surface can be modified with aptamers for increased targeting capacity (Pi

et al., 2018; Wan et al., 2018). Aptamers are oligonucleotide or peptide molecules that have high

affinity and specificity for their targets, and are commonly viewed as alternatives to antibodies for

targeting delivery (Chu et al., 2006). EVs functionalized with necleolin-specific AS1411 aptamer,

produced by simple extrusion of targeting ligands conjugated cells, exhibited enhanced targeting

capacity, improved therapeutic efficacy, and low systemic toxicity in vivo. In a follow-up study,

ACCEPTED MANUSCRIPT

Page 20: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

19

EVs were effectively equipped with prostate-specific membrane antigen aptamer and EGFR

aptamer by simply incubating cholesterol-conjugated RNA aptamers with EVs. Both

reprogrammed-EVs displayed enhanced cancer-cell-specific targeting and efficient suppression of

tumor growth in prostate cancer xenograft and orthotopic breast cancer models, respectively (Pi et

al., 2018) .

An alternative decoration approach to enhance EVs’ tissue targetability is to modify them

with specific chemical compounds. For example, FA and other vitamin receptors are

overexpressed in many cancers of epithelial origin (e.g. colorectal cancer), and have been widely

employed to achieve tumor targeting (Munagala et al., 2016; Zhang et al., 2017). Recently,

Munagala et al. confirmed the feasibility of using drug loaded milk EVs functionalized with FA

for enhanced anti-tumor activity. FA-tagged EV formulations loaded with WFA exhibited

significant higher growth inhibition (74%) than unmodified EV formulations (50%) in mice

bearing human lung cancer (A549) xenografts. Furthermore, such a higher anti-tumor efficacy was

achieved by oral administration, suggesting the potential of effective oral anti-tumor therapy via

functionalized EV-based nanodelivery systems (Munagala et al., 2016). In another study, EVs

were modified with cationized pullulan, a polysaccharide with the ability to target hepatocyte

asialoglycoprotein receptors, which demonstrated not only higher targeting capacity, but also

enhanced therapeutic effect on liver injury (Tamura et al., 2017). Additionally, modification of

Paclitaxel-loaded EVs with AA-PEG moiety, a ligand with high affinity for sigma receptor

(Banerjee et al., 2004), led to greater accumulation of paclitaxel to lung cancer cells that

overexpressed with sigma receptor (Kim et al., 2018). Taken together, surface modification of EVs

may serve as a promising approach to enhance the targeting ability of these vesicles to desired

tissues and improve therapeutic efficacy.

Other than ligand modification, tissue-specific delivery may also be achieved by loading EVs

with magnetic nanoparticles. In a proof-of-concept study by Silva et al., macrophages were

encapsulated with therapeutic agents together with iron oxide nanoparticles. Consequently, EVs

loaded with therapeutics and magnetic nanoparticles were harvested. Magnetic EVs showed

enhanced targeting capacity and greater antitumor effects (Silva et al., 2015). More recently, Qi et

al. suggested superparamagnetic magnetite colloidal nanocrystal clusters (SMCNCs) was also

promising to endow EVs with magnetic-targeting properties. SMCN modified EVs were obtained

ACCEPTED MANUSCRIPT

Page 21: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

20

by incubating mouse serum with transferring-conjugated SMCNCs and harvesting EVs with

superparamagnetic characteristics via magnetic separation. By applying external magnetic field,

SMCN-positive EVs loaded doxorubicin displayed greater accumulation at tumor sites, and

thereby resulting in greater suppression of tumor growth, compared with doxorubicin alone (Qi et

al., 2016). In another study, EV producing cells were first incubated with biotin-functionalized 1,

2-distearoyl-sn-glycero-3- phosphoethanolamine (DSPE)-PEG and FA-functionalized DSPE-PEG.

Then the magnetic and FA-functionalized EVs were obtained by conjugating FA/biotin-EVs with

SA-IONPs and recovered by magnetic separation. Magnetic and FA-functionalized EVs were

endowed with targeting properties and showed significantly enhanced antitumor efficacy both in

vitro and in vivo (Zhang et al., 2017).

4. 3. Modified EVs for enhanced cytoplasmic delivery efficiency

For efficient cytoplasmic delivery of EVs, a deep insight into their uptake mechanisms is

required. Generally, EVs gain entry into cells by either membrane fusion or endocytosis pathways

(Mulcahy et al., 2014). To be specific, there are a variety of endocytic pathways, including

clathrin-dependent endocytosis, and clathrin-independent pathways, such as caveolin-mediated

internalization, macropinocytosis, and lipid raft-mediated uptake (Mulcahy et al., 2014). In

addition, EVs uptake is likely to occur via more than one route, given that failures to completely

abrogate EV internalization upon the treatment with numerous inhibitors were frequently observed

(Barrã¨S et al., 2010; Escrevente et al., 2011; Morelli et al., 2004). For functional delivery of EV

contents into recipient cells, vesicles require in some way to fuse with cell membrane, either direct

at the plasma membrane or fusion with the limited endosomal membranes after cellular uptake

(Stranford and Leonard, 2017). EVs derived from specific cell types are capable of fusing with

target cells due to the expression of specific membrane proteins. For instance, it was reported that

EVs from DCs could bypass the endosomal-lysosomal pathway by directly fusing with the plasma

membrane through their tetraspanin CD9 interacting with surface glycoproteins on the target cells,

thus offering an efficient pathway for cytoplasmic delivery (Montecalvo et al., 2012; Van den

Boorn et al., 2011). However, the cellular uptake of EVs secreted by most cells is accomplished by

endocytosis (Morelli et al., 2004; Thery et al., 2009; Tian et al., 2010), which is considered

unsatisfactory for therapeutic use (Nakase and Futaki, 2015). First, there is a fierce competition

between the exogenously administered EVs and large number of endogenous EVs presented in the

ACCEPTED MANUSCRIPT

Page 22: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

21

body fluids (e.g. approximately 3,000,000 EVs per microliter in the blood) (Vlassov et al., 2012).

Besides, the low cellular uptake efficiency of EVs may be partly due to the charge repulsion

existing between EVs with negatively charged cell membranes (Nakase et al., 2015). Furthermore,

when EVs are taken up by recipient cells and trafficked in the endosomal/lysosomal compartments,

EV cargo may suffer from acidification and digestion during the process of endosomal maturation

(Mindell, 2012). As a result, this may reduce the biological activity of EV’s contents before their

release to the cytosol. In fact, inefficient escape from endosomes into the cytosol represents one of

the major obstacles for intracellular targeting of EVs, which is similar to synthetic nanomaterials

(Morishita et al., 2017; Nakase and Futaki, 2015). Therefore, an important consideration for

EV-based delivery vehicles is to endow them with a functionality to breach the endosomal

membrane for the cytoplasmic release of their cargoes.

Although, a number of strategies have been reported to arm EVs with targeting properties,

approaches to enhance the cellular uptake of EVs and promote cytosolic release of their payloads

are relatively limited (Table 4). Nevertheless, several encouraging results have been recently

reported. Owing to the similarities in composition, function, and biogenesis process between EVs

and viruses (Nolte-'t Hoen et al., 2016), it has been suggested that incorporation of viral

components to EV membrane may be a useful tool to improve the intracellular delivery efficiency

of EVs (Koppers-Lalic et al., 2013). For example, by taking advantages of the pH-sensitive

properties of G protein of vascular stomatitis virus (VSV-G), Yang et al. engineered EVs to

express VSV-G on the surface to promote direct membrane fusion with recipient cells under acidic

conditions (e.g. in muscle tissues). As a result, VSV-G modified EVs facilitated direct membrane

fusion with target cells and delivery of biologically active membrane protein glucose transporter-4

directly into the cell membrane of recipient cell (Fig. 3) (Yang et al., 2017c). Of note, in addition

to transfer of membrane protein, VSV-G functionalized EVs may be readily applicable to deliver

encapsulated payloads, such as small chemotherapeutics, RNAs, and proteins, to the cytosol of

target cells, bypassing endocytic trafficking. An attractive strategy to promote endosomal escape

via membrane fusion at low pH is to use a pH-sensitive fusogenic GALA peptide. In a recent study,

Nakase et al. developed an efficient method to enhance the cellular uptake and cytosolic release of

EVs by combining cationic lipids with GALA peptide. Cationic lipids that acted as a “glue” to

facilitate cellular uptake by mitigating charge repulsion between EVs and recipient cells, while

ACCEPTED MANUSCRIPT

Page 23: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

22

GALA peptide mediated membrane fusion of endosome and EVs at low pH, and promoted

cytosolic release of encapsulated ribosome-inactivating protein saporin (Fig. 3) (Nakase and

Futaki, 2015). Similarly, display of GALA peptide on the surface of EVs was also employed to

enhance tumor antigen presentation capacity for improved antitumor therapy (Morishita et al.,

2017).

An alternative strategy to improve EVs’ cytoplasmic delivery efficiency is to activate

macropinocytosis. Induction of macropinocytosis is accompanied by actin reorganization, ruffling

of plasma membrane, and engulfment of large volumes of extracellular fluid, which could lead to

the enhancement in cellular EV uptake efficiency (Nakase et al., 2015). With the intrinsic ability

to translocate into most cells both in vitro and in vivo, CPPs have been frequently used to enhance

the cellular uptake of nanoparticles (Tiwari et al., 2014; van Asbeck et al., 2013). Recently,

El-Andaloussi has reported an approach to incorporate CPPs on EV surface via fusion with

Lamp2b protein to enhance cellular translocation of siRNA (Elandaloussi et al., 2012). More

recently, Nakase and coworkers suggested that displaying arginine-rich CPPs on EV surface was

able to induce macropinocytosis and boost effective cellular uptake of EVs (Fig. 3) (Nakase et al.,

2016). Furthermore, the number of arginine residues in the peptide sequences could affect the

cellular EV uptake efficiency. Hexadeca-arginine peptide (R16) modified EVs loaded with saporin

showed effective anticancer activity, possibly due to the higher endosomal membrane perturbation

ability of R16 than that of shorter oligoarginines (Nakase et al., 2017). Still, as the authors

presented, further research is needed to elucidate the scaffold effects on the functionality of CPPs

other than the number of arginine residues (Nakase et al., 2017). Recently, Akishiba developed a

novel endosomolytic peptide (L17E) by introducing one glutamic acid residues into the

hydrophobic face. L17E possessed two distinct characteristics, including preferential disruption of

negatively charged membranes (endosomal membranes) over slightly less negatively charged

plasma membranes, and promotion of cellular uptake by activating macropinocytosis (Akishiba et

al., 2017). The author further demonstrated that L17E functionalized EVs showed effectiveness in

cytoplasmic delivery of EV encapsulated dextran (Akishiba et al., 2017).

5. Conclusion

The discovery that EVs are capable of transferring biomolecules between cells and altering

the recipient cells' phenotype enables them to be attractive candidates for drug delivery vehicles.

ACCEPTED MANUSCRIPT

Page 24: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

23

EVs may possess several advantages over conventional drug delivery systems, such as high

biocompatibility and safety, low immunogenicity, and ability to traverse biological barriers.

However, the rapid clearance of unmodified exogenously administered EVs highlights the need to

protect them from the recognition by MPS and protract their circulation half-life. Besides, in order

to achieve targeted delivery, their intrinsic targeting capacity should be further tuned. Furthermore,

the efficiency of EVs to overcome biological barriers for reaching intracellular targets, still

remains unsatisfactory. In this context, for therapeutic applications, EVs need to be engineered to

prolong their circulation time, improve targeting properties, and to enhance cytoplasmic delivery

efficiency. Considering that multiple EVs’ biological interaction occurs through surface interaction,

surface engineering is fundamental for the development of advanced EV-based nanodelivery

systems. “Cloacking” EVs with hydrophilic moieties, such as PEG derivatives, has been proven to

confer these vesicles with stealth properties, allowing them to evade clearance by the immune

system. The incorporation of targeting ligands to EVs imparts them with enhanced targeting

properties, enabling a greater accumulation at target site and reducing off-targets effects on

healthy tissues. Display of VSV-G, GALA or other functional ligands on EV surface, has been

demonstrated to be attractive tools to boost the cellular uptake and cytosolic delivery of their

payloads. Together, functionalized EVs, offering the tantalizing prospect of extending the delivery

capability of native vesicles, may represent the next generation of therapeutic nanodelivery

vehicles that combines the biological features of EVs, such as high biocompatibility and low

immunogenicity, with the enhanced delivery properties of functional moieties. For EV

modification, “cell engineering” enables display of ligands with predictable orientation on EV

surface, however, may possess complexity, inefficiency, and long stability issues, (Richards et al.,

2016). While direct “EV engineering” allows for a controllable and efficient incorporation of

various ligands, but pose possible risk of damaging EVs’ structural and biological integrity. As

each modification techniques has certain benefits and drawbacks, when selecting a modification

approach, it is essential to consider the complexity of the modification systems, the properties of

functional ligands, and downstream applications (Armstrong et al., 2017). To this end, there is a

continuous need to develop novel modification strategies to efficiently maximize the potential of

modified EVs in therapeutic delivery. As an example, liposome-based cellular engineering has

been proven to be capable of introducing multiple ligands of divergent functionality (Lee et al.,

ACCEPTED MANUSCRIPT

Page 25: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

24

2016). This is quite fascinating as EVs could be simultaneously functionalized with

immune-evading, targeting, and uptake-enhancing ligands to prolong their circulation half-life,

targeting capacity, and cytoplasmic delivery efficiency.

However, several challenges remain to reach maximum potential of modified EVs in clinic.

First, it warrants thorough investigation dedicated towards assessing the potential effects of

modification techniques on the biological functionality of EVs. This is because manipulation of

EV surface may alter the proper orientation of EVs’ membrane proteins, and expose certain

antigens that could be recognize by the immune system (Mout et al., 2012). This in turn can be a

potential source of immunogenic reactions that may cause immune responses on potential patients,

especially during chronic treatments. Modification process also bares the risk of damaging the

functionality of ligands. In this circumstance, a crucial concern is to distinguish between cellular

uptake and functional delivery. As when the functional ligands on vesicle surface are inactivated,

EVs can still be internalized by recipient cells via nonspecific endocytosis. However, inactivated

ligands would be unable to trigger specific signal cascades. For example, EVs may be unable to

fuse with target cells and release their payloads to the desired compartment of recipient cell when

the ligands on their surface are inactivated (Hung and Leonard, 2016). Therefore, the first step for

modified EVs to reach clinical use is the assessment of their functionality and potential

immunogenicity. Additionally, effective modification of EVs and harnessing them for therapeutic

delivery highlights the need for gaining deeper insight into EV biology. EVs are nonliving entities,

which may benefit modifications under harsher modification conditions compared with cells.

However, the significant smaller particle size and more rigid membrane of EVs as compared with

cells would possibly make it more complex for surface engineering (Parolini et al., 2009). Hence,

addressing our knowledge gaps is critical for empowering continued innovation for developing

novel modification techniques. Lastly, exploring more functional moieties may considerably assist

in improving the delivery properties and broadening the therapeutic applications of EVs. Ideal

candidates may possess several features, such as competent functionality, small molecular weight,

high chemical and thermal stability, and facile selection and production protocols (Revets et al.,

2005). With the use of these moieties, it would, therefore, be possible to achieve higher circulation

stability, greater tissue specificity, and improved delivery efficiency of EVs following systemic

administration, which may enhance the bioavailability and therapeutic efficacy of EV payloads.

ACCEPTED MANUSCRIPT

Page 26: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

25

Together, resolution of these challenges through energetic collaboration of scientists from different

disciplines could minimize the future bottlenecks in the development of novel functionalization

strategies and boost these functionalized nanovesicles towards efficient and safe drug delivery in

clinic.

Conflict of interest:

The authors declare no conflict of interests relevant to this work.

Acknowledgements:

The authors are grateful for the financial support from the National Natural Science

Foundation of China (Grant No. 81373335).

References

Akishiba, M., Takeuchi, T., Kawaguchi, Y., Sakamoto, K., Yu, H.H., Nakase, I., Takataninakase,

T., Madani, F., Gräslund, A., Futaki, S., 2017. Cytosolic antibody delivery by lipid-sensitive

endosomolytic peptide. Nat. Chem. 9, 751-761.

Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., Wood, M.J.A., 2011. Delivery of

siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29,

341-345.

Amoozgar, Z., Yeo, Y., 2012. Recent advances in stealth coating of nanoparticle drug delivery

systems. Wiley Interdiscip. Rev.-Nanomed. Nanobiotechnol. 4, 219-233.

Andreu, Z., Oshiro, R.O., Redruello, A., Lopez-Martin, S., Gutierrez-Vazquez, C., Morato, E.,

Marina, A.I., Gomez, C.O., Yanez-Mo, M., 2017. Extracellular vesicles as a source for

non-invasive biomarkers in bladder cancer progression. Eur. J. Pharm. Sci. 98, 70-79.

Armstrong, J.P.K., Holme, M.N., Stevens, M.M., 2017. Re-engineering extracellular vesicles as

smart nanoscale therapeutics. ACS Nano 11, 69-83.

Baj-Krzyworzeka, M., Szatanek, R., Weglarczyk, K., Baran, J., Zembala, M., 2007.

Tumour-derived microvesicles modulate biological activity of human monocytes. Immunol. Lett.

113, 76-82.

Banerjee, R., Tyagi, P., Li, S., Huang, L., 2004. Anisamide-targeted stealth liposomes: A potent

carrier for targeting doxorubicin to human prostate cancer cells. Int. J. Cancer 112, 693-700.

Barile, L., Vassalli, G., 2017. Exosomes: Therapy delivery tools and biomarkers of diseases.

Pharmacol. Ther. 174, 63-78.

Barrã¨S, C., Blanc, L., Bette-Bobillo, P., Andrã©, S., Mamoun, R., Gabius, H.J., Vidal, M., 2010.

Galectin-5 is bound onto the surface of rat reticulocyte exosomes and modulates vesicle uptake by

macrophages. Blood 115, 696-705.

Bellavia, D., Raimondo, S., Calabrese, G., Forte, S., Cristaldi, M., Patinella, A., Memeo, L.,

Manno, M., Raccosta, S., Diana, P., Cirrincione, G., Giavaresi, G., Monteleone, F., Fontana, S., De

ACCEPTED MANUSCRIPT

Page 27: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

26

Leo, G., Alessandro, R., 2017. Interleukin 3-receptor targeted exosomes inhibit in vitro and in vivo

Chronic Myelogenous Leukemia cell growth. Theranostics 7, 1333-1345.

Bryniarski, K., Ptak, W., Jayakumar, A., Puellmann, K., Caplan, M.J., Chairoungdua, A., Lu, J.,

Adams, B.D., Sikora, E., Nazimek, K., Marquez, S., Kleinstein, S.H., Sangwung, P., Iwakiri, Y.,

Delgato, E., Redegeld, F., Blokhuis, B.R., Wojcikowski, J., Daniel, A.W., Kormelink, T.G.,

Askenase, P.W., 2013. Antigen-specific, antibody-coated, exosome-like nanovesicles deliver

suppressor T-cell microRNA-150 to effector T cells to inhibit contact sensitivity. J. Allergy Clin.

Immunol. 132, 170-181.

Cappello, F., Logozzi, M., Campanella, C., Bavisotto, C.C., Marcilla, A., Properzi, F., Fais, S.,

2017. Exosome levels in human body fluids: A tumor marker by themselves? Eur. J. Pharm. Sci.

96, 93-98.

Caracciolo, G., 2015. Liposome–protein corona in a physiological environment: Challenges and

opportunities for targeted delivery of nanomedicines. Nanomedicine 11, 543-557.

Chu, T.C., Twu, K.Y., Ellington, A.D., Levy, M., 2006. Aptamer mediated siRNA delivery.

Nucleic Acids Res. 34, e73.

Clavijo-Jordan, V., Kodibagkar, V.D., Beeman, S.C., Hann, B.D., Bennett, K.M., 2012. Principles

and emerging applications of nanomagnetic materials in medicine. Wiley Interdiscip.

Rev.-Nanomed. Nanobiotechnol. 4, 345-365.

Clayton, A., Harris, C.L., Court, J., Mason, M.D., Morgan, B.P., 2003. Antigen-presenting cell

exosomes are protected from complement-mediated lysis by expression of CD55 and CD59. Eur. J.

Immunol. 33, 522-531.

Colombo, M., Raposo, G., Thery, C., 2014. Biogenesis, secretion, and intercellular interactions of

exosomes and other extracellular vesicles. Annu. Rev. Cell and Dev. Bi. 30, 255-289.

Delcayre, A., Estelles, A., Sperinde, J., Roulon, T., Paz, P., Aguilar, B., Villanueva, J., Khine, S.,

Le Pecq, J.B., 2005. Exosome display technology: Applications to the development of new

diagnostics and therapeutics. Blood Cell. Mol. and Dis. 35, 158-168.

Deschout, H., Raemdonck, K., Stremersch, S., Maoddi, P., Mernier, G., Renaud, P., Jiguet, S.,

Hendrix, A., Bracke, M., Van den Broecke, R., Roeding, M., Rudemo, M., Demeester, J., De

Smedt, S.C., Strubbe, F., Neyts, K., Braeckmans, K., 2014. On-chip light sheet illumination

enables diagnostic size and concentration measurements of membrane vesicles in biofluids.

Nanoscale 6, 1741-1747.

Duncan, R., 2003. The dawning era of polymer therapeutics. Nat. Rev. Drug Discov. 2, 347-360.

El Andaloussi, S., Maeger, I., Breakefield, X.O., Wood, M.J.A., 2013. Extracellular vesicles:

biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 348-358.

Elandaloussi, S., Yi, L., Lakhallittleton, S., Li, J., Seow, Y., Gardiner, C., Alvarezerviti, L., Sargent,

I.L., Wood, M.J.A., 2012. Exosome-mediated delivery of siRNA in vitro and in vivo. Nat. Protoc.

7, 2112-2126.

Escrevente, C., Keller, S., Altevogt, P., Costa, J., 2011. Interaction and uptake of exosomes by

ovarian cancer cells. BMC Cancer 11, 108.

Fais, S., O'Driscoll, L., Borras, F.E., Buzas, E., Camussi, G., Cappello, F., Carvalho, J., da Silva,

A.C., Del Portillo, H., El Andaloussi, S., Trcek, T.F., Furlan, R., Hendrix, A., Gursel, I., Kralj-Iglic,

V., Kaeffer, B., Kosanovic, M., Lekka, M.E., Lipps, G., Logozzi, M., Marcilla, A., Sammar, M.,

Llorente, A., Nazarenko, I., Oliveira, C., Pocsfalvi, G., Rajendran, L., Raposo, G., Rohde, E.,

Siljander, P., van Niel, G., Vasconcelos, M.H., Yanez-Mo, M., Yliperttula, M.L., Zarovni, N.,

ACCEPTED MANUSCRIPT

Page 28: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

27

Zavec, A.B., Giebel, B., 2016. Evidence-based clinical use of nanoscale extracellular vesicles in

nanomedicine. Acs Nano 10, 3886-3899.

Fais, S., Venturi, G., Gatenby, B., 2014. Microenvironmental acidosis in carcinogenesis and

metastases: new strategies in prevention and therapy. Cancer Metast. Rev. 33, 1095-1108.

Federici, C., Petrucci, F., Caimi, S., Cesolini, A., Logozzi, M., Borghi, M., D'Ilio, S., Lugini, L.,

Violante, N., Azzarito, T., Majorani, C., Brambilla, D., Fais, S., 2014. Exosome release and low

pH belong to a framework of resistance of human melanoma cells to cisplatin. Plos One 9,

e88193.

Feng, D., Zhao, W.L., Ye, Y.Y., Bai, X.C., Liu, R.Q., Chang, L.F., Zhou, Q., Sui, S.F., 2010.

Cellular internalization of exosomes occurs through phagocytosis. Traffic 11, 675-687.

Fuhrmann, G., Herrmann, I.K., Stevens, M.M., 2015. Cell-derived vesicles for drug therapy and

diagnostics: Opportunities and challenges. Nano Today 10, 397-409.

Gujrati, V., Kim, S., Kim, S.H., Min, J.J., Choy, H.E., Kim, S.C., Jon, S., 2014. Bioengineered

Bacterial outer membrane vesicles as cell-specific drug-delivery vehicles for cancer therapy. ACS

Nano 8, 1525-1537.

Hadla, M., Palazzolo, S., Corona, G., Caligiuri, I., Canzonieri, V., Toffoli, G., Rizzolio, F., 2016.

Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse

models. Nanomedicine 11, 2431-2441.

Haraszti, R.A., Didiot, M.-C., Sapp, E., Leszyk, J., Shaffer, S.A., Rockwell, H.E., Gao, F., Narain,

N.R., DiFiglia, M., Kiebish, M.A., Aronin, N., Khvorova, A., 2016. High-resolution proteomic

and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracellular

Vesicles 5, 32570.

Hartman, Z.C., Wei, J., Glass, O.K., Guo, H., Lei, G., Yang, X.Y., Osada, T., Hobeika, A.,

Delcayre, A., Le, P.J., 2011. Increasing vaccine potency through exosome antigen targeting.

Vaccine 29, 9361-9367.

Haussecker, D., 2014. Current issues of RNAi therapeutics delivery and development. J. Control.

Release 195, 49-54.

Hung, M.E., Leonard, J.N., 2015. Stabilization of exosome-targeting peptides via engineered

glycosylation. J. Biol. Chem. 290, 8166-8172.

Hung, M.E., Leonard, J.N., 2016. A platform for actively loading cargo RNA to elucidate limiting

steps in EV-mediated delivery. J. Extracellular Vesicles 5, 31027.

Iessi, E., Logozzi, M., Lugini, L., Azzarito, T., Federici, C., Spugnini, E.P., Mizzoni, D., Di Raimo,

R., Angelini, D.F., Battistini, L., Cecchetti, S., Fais, S., 2017. Acridine Orange/exosomes increase

the delivery and the effectiveness of Acridine Orange in human melanoma cells: A new prototype

for theranostics of tumors. J. Enzym. Inhib. Med. Chem. 32, 648-657.

Imai, T., Takahashi, Y., Nishikawa, M., Kato, K., Morishita, M., Yamashita, T., Matsumoto, A.,

Charoenviriyakul, C., Takakura, Y., 2015. Macrophage-dependent clearance of systemically

administered B16BL6-derived exosomes from the blood circulation in mice. J. Extracellular

Vesicles 4, 26238.

Ishida, T., Ichihara, M., Wang, X., Yamamoto, K., Kimura, J., Majima, E., Kiwada, H., 2006.

Injection of PEGylated liposomes in rats elicits PEG-specific IgM, which is responsible for rapid

elimination of a second dose of PEGylated liposomes. J. Control. Release 112, 15-25.

Jiang, L., Vader, P., Schiffelers, R.M., 2017. Extracellular vesicles for nucleic acid delivery:

progress and prospects for safe RNA-based gene therapy. Gene Ther. 24, 157-166.

ACCEPTED MANUSCRIPT

Page 29: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

28

Johnsen, K.B., Gudbergsson, J.M., Skov, M.N., Pilgaard, L., Moos, T., Duroux, M., 2014. A

comprehensive overview of exosomes as drug delivery vehicles - endogenous nanocarriers for

targeted cancer therapy. Biochim. Biophys. Acta 1846, 75-87.

Kamerkar, S., LeBleu, V.S., Sugimoto, H., Yang, S., Ruivo, C.F., Melo, S.A., Lee, J.J., Kalluri, R.,

2017. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature

546, 498.

Katakowski, M., Buller, B., Zheng, X., Lu, Y., Rogers, T., Osobamiro, O., Shu, W., Jiang, F.,

Chopp, M., 2013. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma

growth. Cancer Lett. 335, 201-204.

Kim, M.S., Haney, M.J., Zhao, Y., Yuan, D., Deygen, I., Klyachko, N.L., Kabanov, A.V.,

Batrakova, E.V., 2018. Engineering macrophage-derived exosomes for targeted paclitaxel delivery

to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine 14, 195-204.

Kim, T.K., Eberwine, J.H., 2010. Mammalian cell transfection: the present and the future. Anal.

Bioanal. Chem. 397, 3173-3178.

Klutz, K., Schaffert, D., Willhauck, M.J., Gruenwald, G.K., Haase, R., Wunderlich, N., Zach, C.,

Gildehaus, F.J., Senekowitsch-Schmidtke, R., Goeke, B., Wagner, E., Ogris, M., Spitzweg, C.,

2011. Epidermal growth factor receptor-targeted I-131-therapy of liver cancer following systemic

delivery of the sodium iodide symporter gene. Mol. Ther. 19, 676-685.

Koh, E., Lee, E.J., Nam, G.-H., Hong, Y., Cho, E., Yang, Y., Kim, I.-S., 2017. Exosome-SIRP

alpha, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 121, 121-129.

Kooijmans, S.A.A., Aleza, C.G., Roffler, S.R., van Solinge, W.W., Vader, P., Schiffelers, R.M.,

2016a. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour

cell targeting. J. Extracellular Vesicles 5, 31053.

Kooijmans, S.A.A., Fliervoet, L.A.L., van der Meel, R., Fens, M.H.A.M., Heijnen, H.F.G.,

Henegouwen, P.M.P.v.B.E., Vader, P., Schiffelers, R.M., 2016b. PEGylated and targeted

extracellular vesicles display enhanced cell specificity and circulation time. J. Control. Release

224, 77-85.

Kooijmans, S.A.A., Gitz-Francois, J., Schiffelers, R.M., Vader, P., 2018. Recombinant

phosphatidylserine-binding nanobodies for targeting of extracellular vesicles to tumor cells: a

plug-and-play approach. Nanoscale 10, 2413-2426.

Kooijmans, S.A.A., Schiffelers, R.M., Zarovni, N., Vago, R., 2016c. Modulation of tissue tropism

and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer

treatment. Pharmacol. Res. 111, 487-500.

Koppers-Lalic, D., Hogenboom, M.M., Middeldorp, J.M., Pegtel, D.M., 2013. Virus-modified

exosomes for targeted RNA delivery; A new approach in nanomedicine. Adv. Drug Deliv. Rev. 65,

348-356.

Kumari, A., Yadav, S.K., Yadav, S.C., 2010. Biodegradable polymeric nanoparticles based drug

delivery systems. Colloid Surf. B-Biointerfaces 75, 1-18.

Kusuzaki, K., Matsubara, T., Murata, H., Logozzi, M., Iessi, E., Di Raimo, R., Carta, F., Supuran,

C.T., Fais, S., 2017. Natural extracellular nanovesicles and photodynamic molecules: is there a

future for drug delivery? J. Enzym. Inhib. Med. Chem. 32, 908-916.

Lai, C.P., Kim, E.Y., Badr, C.E., Weissleder, R., Mempel, T.R., Tannous, B.A., Breakefield, X.O.,

2015. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation

using multiplexed reporters. Nat. Commun. 6, 7029.

ACCEPTED MANUSCRIPT

Page 30: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

29

Lai, C.P., Mardini, O., Ericsson, M., Prabhakar, S., Maguire, C.A., Chen, J.W., Tannous, B.A.,

Breakefield, X.O., 2014. Dynamic biodistribution of extracellular vesicles in vivo using a

multimodal imaging reporter. Acs Nano 8, 483-494.

Lai, R.C., Yeo, R.W., Tan, K.H., Lim, S.K., 2013. Exosomes for drug delivery - a novel

application for the mesenchymal stem cell. Biotechnol. Adv. 31, 543-551.

Lamichhane, T.N., Raiker, R.S., Jay, S.M., 2015. Exogenous DNA loading into extracellular

vesicles via electroporation is size-dependent and enables limited gene delivery. Mol. Pharm. 12,

3650-3657.

Lee, J., Lee, H., Goh, U., Kim, J., Jeong, M., Lee, J., Park, J.H., 2016. Cellular engineering with

membrane fusogenic liposomes to produce functionalized extracellular vesicles. ACS Appl. Mater.

Interfaces 8, 6790-6795.

Lener, T., Gimona, M., Aigner, L., Borger, V., Buzas, E., Camussi, G., Chaput, N., Chatterjee, D.,

Court, F.A., Del Portillo, H.A., O'Driscoll, L., Fais, S., Falcon-Perez, J.M., Felderhoff-Mueser, U.,

Fraile, L., Gho, Y.S., Gorgens, A., Gupta, R.C., Hendrix, A., Hermann, D.M., Hill, A.F., Hochberg,

F., Horn, P.A., de Kleijn, D., Kordelas, L., Kramer, B.W., Kramer-Albers, E.-M.,

Laner-Plamberger, S., Laitinen, S., Leonardi, T., Lorenowicz, M.J., Lim, S.K., Lotvall, J., Maguire,

C.A., Marcilla, A., Nazarenko, I., Ochiya, T., Patel, T., Pedersen, S., Pocsfalvi, G., Pluchino, S.,

Quesenberry, P., Reischl, I.G., Rivera, F.J., Sanzenbacher, R., Schallmoser, K., Slaper-Cortenbach,

I., Strunk, D., Tonn, T., Vader, P., van Balkom, B.W.M., Wauben, M., Andaloussi, S.E., Thery, C.,

Rohde, E., Giebel, B., 2015. Applying extracellular vesicles based therapeutics in clinical trials -

an ISEV position paper. J. Extracellular Vesicles 4, 30087.

Liu, Y., Li, D., Liu, Z., Zhou, Y., Chu, D., Li, X., Jiang, X., Hou, D., Chen, X., Chen, Y., 2015.

Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine

relapse. Sci. Rep. 5, 17543.

Logozzi, M., Angelini, D.F., Iessi, E., Mizzoni, D., Di Raimo, R., Federici, C., Lugini, L.,

Borsellino, G., Gentilucci, A., Pierella, F., Marzio, V., Sciarra, A., Battistini, L., Fais, S., 2017.

Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients.

Cancer Lett. 403, 318-329.

Logozzi, M., De Milito, A., Lugini, L., Borghi, M., Calabro, L., Spada, M., Perdicchio, M.,

Marino, M.L., Federici, C., Iessi, E., Brambilla, D., Venturi, G., Lozupone, F., Santinami, M.,

Huber, V., Maio, M., Rivoltini, L., Fais, S., 2009. High levels of exosomes expressing CD63 and

Caveolin-1 in plasma of melanoma patients. Plos One 4, e5219.

Long, K.B., Beatty, G.L., 2013. Harnessing the antitumor potential of macrophages for cancer

immunotherapy. OncoImmunology 2, e26860.

Lu, M., Xing, H., Yang, Z., Sun, Y., Yang, T., Zhao, X., Cai, C., Wang, D., Ding, P., 2017. Recent

advances on extracellular vesicles in therapeutic delivery: Challenges, solutions, and opportunities.

Eur. J. Pharm. Biopharm. 119, 381-395.

Martins-Marques, T., Pinho, M.J., Zuzarte, M., Oliveira, C., Pereira, P., Sluijter, J.P.G., Gomes, C.,

Girao, H., 2016. Presence of Cx43 in extracellular vesicles reduces the cardiotoxicity of the

anti-tumour therapeutic approach with doxorubicin. J. Extracellular Vesicles 5, 32538.

Mindell, J.A., 2012. Lysosomal acidification mechanisms. Annu. Rev. Physiol. 74, 69-86.

Momen-Heravi, F., Bala, S., Bukong, T., Szabo, G., 2014. Exosome-mediated delivery of

functionally active miRNA-155 inhibitor to macrophages. Nanomedicine 10, 1517-1527.

Montecalvo, A., Larregina, A.T., Shufesky, W.J., Stolz, D.B., Sullivan, M.L.G., Karlsson, J.M.,

ACCEPTED MANUSCRIPT

Page 31: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

30

Baty, C.J., Gibson, G.A., Erdos, G., Wang, Z.L., Milosevic, J., Tkacheva, O.A., Divito, S.J., Jordan,

R., Lyons-Weiler, J., Watkins, S.C., Morelli, A.E., 2012. Mechanism of transfer of functional

microRNAs between mouse dendritic cells via exosomes. Blood 119, 756-766.

Morelli, A.E., Larregina, A.T., Shufesky, W.J., Sullivan, M.L.G., Stolz, D.B., Papworth, G.D.,

Zahorchak, A.F., Logar, A.J., Wang, Z., Watkins, S.C., 2004. Endocytosis, intracellular sorting,

and processing of exosomes by dendritic cells. Blood 104, 3257-3266.

Morishita, M., Takahashi, Y., Nishikawa, M., Ariizumi, R., Takakura, Y., 2017. Enhanced class I

tumor antigen presentation via cytosolic delivery of exosomal cargos by tumor cell-derived

exosomes displaying a pH-sensitive fusogenic peptide. Mol. Pharm. 14, 4079-4086.

Morishita, M., Takahashi, Y., Nishikawa, M., Sano, K., Kato, K., Yamashita, T., Imai, T., Saji, H.,

Takakura, Y., 2015. Quantitative analysis of tissue distribution of the B16BL6-derived exosomes

using a streptavidin-lactadherin fusion protein and iodine-125-labeled biotin derivative after

intravenous injection in mice. J. Pharm. Sci. 104, 705-713.

Mout, R., Moyano, D.F., Rana, S., Rotello, V.M., 2012. Surface functionalization of nanoparticles

for nanomedicine. Chem. Soc. Rev. 41, 2539-2544.

Mulcahy, L.A., Pink, R.C., Carter, D.R.F., 2014. Routes and mechanisms of extracellular vesicle

uptake. J. Extracellular Vesicles 3, 24641.

Munagala, R., Aqil, F., Jeyabalan, J., Gupta, R.C., 2016. Bovine milk-derived exosomes for drug

delivery. Cancer Lett. 371, 48-61.

Nakase, I., Futaki, S., 2015. Combined treatment with a pH-sensitive fusogenic peptide and

cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci. Rep. 5, 10112.

Nakase, I., Kobayashi, N.B., Takatani-Nakase, T., Yoshida, T., 2015. Active macropinocytosis

induction by stimulation of epidermal growth factor receptor and oncogenic Ras expression

potentiates cellular uptake efficacy of exosomes. Sci. Rep. 5, 10300.

Nakase, I., Noguchi, K., Aoki, A., Takatani-Nakase, T., Fujii, I., Futaki, S., 2017. Arginine-rich

cell-penetrating peptide-modified extracellular vesicles for active macropinocytosis induction and

efficient intracellular delivery. Sci. Rep. 7, 1991.

Nakase, I., Noguchi, K., Fujii, I., Futaki, S., 2016. Vectorization of biomacromolecules into cells

using extracellular vesicles with enhanced internalization induced by macropinocytosis. Scientific

Reports 6, 34937.

Nievergall, E., Ramshaw, H.S., Yong, A.S.M., Biondo, M., Busfield, S.J., Vairo, G., Lopez, A.F.,

Hughes, T.P., White, D.L., Hiwase, D.K., 2014. Monoclonal antibody targeting of IL-3 receptor

alpha with CSL362 effectively depletes CML progenitor and stem cells. Blood 123, 1218-1228.

Nolte-'t Hoen, E., Cremer, T., Gallo, R.C., Margolis, L.B., 2016. Extracellular vesicles and viruses:

Are they close relatives? Proc. Natl. Acad. Sci. U. S. A. 113, 9155-9161.

Ohno, S., Takanashi, M., Sudo, K., Ueda, S., Ishikawa, A., Matsuyama, N., Fujita, K., Mizutani, T.,

Ohgi, T., Ochiya, T., Gotoh, N., Kuroda, M., 2013. Systemically injected exosomes targeted to

EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185-191.

Parolini, I., Federici, C., Raggi, C., Lugini, L., Palleschi, S., De Milito, A., Coscia, C., Iessi, E.,

Logozzi, M., Molinari, A., Colone, M., Tatti, M., Sargiacomo, M., Fais, S., 2009.

Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 284,

34211-34222.

Perche, F., Torchilin, V.P., 2013. Recent trends in multifunctional liposomal nanocarriers for

enhanced tumor targeting. J. Drug Deliv. 2013, 705265.

ACCEPTED MANUSCRIPT

Page 32: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

31

Pi, F.M., Binzel, D.W., Lee, T.J., Li, Z.F., Sun, M.Y., Rychahou, P., Li, H., Haque, F., Wang, S.Y.,

Croce, C.M., Guo, B., Evers, B.M., Guo, P.X., 2018. Nanoparticle orientation to control RNA

loading and ligand display on extracellular vesicles for cancer regression. Nat. Nanotechnol. 13,

82.

Puri, A., Loomis, K., Smith, B., Lee, J.H., Yavlovich, A., Heldman, E., Blumenthal, R., 2009.

Lipid-based nanoparticles as pharmaceutical drug carriers: From concepts to clinic. Crit. Rev. Ther.

Drug Carrier Syst. 26, 523-580.

Qi, H., Liu, C., Long, L., Ren, Y., Zhang, S., Chang, X., Qian, X., Jia, H., Zhao, J., Sun, J., Hou,

X., Yuan, X., Kang, C., 2016. Blood exosomes endowed with magnetic and targeting properties

for cancer Ttherapy. Acs Nano 10, 3323-3333.

Raemdonck, K., Braeckmans, K., Demeester, J., De Smedt, S.C., 2014. Merging the best of both

worlds: hybrid lipid-enveloped matrix nanocomposites in drug delivery. Chem. Soc. Rev. 43,

444-472.

Raposo, G., Stoorvogel, W., 2013. Extracellular vesicles: Exosomes, microvesicles, and friends. J.

Cell Biol. 200, 373-383.

Revets, H., De, B.P., Muyldermans, S., 2005. Nanobodies as novel agents for cancer therapy.

Expert Opin. Biol. Ther. 5, 111-124.

Richards, M.J., Hsia, C.Y., Singh, R.R., Haider, S.H., Kumpf, J.M., Kawate, T., Daniel, S., 2016.

Membrane protein mobility and orientation preserved in supported bilayers created directly from

cell plasma membrane blebs. Langmuir 32, 2963-2974.

Saari, H., Lazaro-Ibanez, E., Viitala, T., Vuorimaa-Laukkanen, E., Siljander, P., Yliperttula, M.,

2015. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of Paclitaxel

in autologous prostate cancer cells. J. Control. Release 220, 727-737.

Saunderson, S.C., Dunn, A.C., Crocker, P.R., McLellan, A.D., 2014. CD169 mediates the capture

of exosomes in spleen and lymph node. Blood 123, 208-216.

Silva, A.K.A., Kolosnjaj-Tabi, J., Bonneau, S., Marangon, I., Boggetto, N., Aubertin, K., Clement,

O., Bureau, M.F., Luciani, N., Gazeau, F., Wilhelm, C., 2013. Magnetic and photoresponsive

theranosomes: translating cell-released vesicles into smart nanovectors for cancer therapy. Acs

Nano 7, 4954-4966.

Silva, A.K.A., Luciani, N., Gazeau, F., Aubertin, K., Bonneau, S., Chauvierre, C., Letourneur, D.,

Wilhelm, C., 2015. Combining magnetic nanoparticles with cell derived microvesicles for drug

loading and targeting. Nanomedicine 11, 645-655.

Smyth, T., Kullberg, M., Malik, N., Smithjones, P., Graner, M.W., Anchordoquy, T.J., 2015.

Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J. Control.

Release 199, 145-155.

Smyth, T., Petrova, K., Payton, N.M., Persaud, I., Redzic, J.S., Gruner, M.W., Smith-Jones, P.,

Anchordoquy, T.J., 2014. Surface functionalization of exosomes using click chemistry. Bioconjug.

Chem. 25, 1777-1784.

Spugnini, E., Fais, S., 2017. Proton pump inhibition and cancer therapeutics: A specific tumor

targeting or it is a phenomenon secondary to a systemic buffering? Semin. Cancer Biol. 43,

111-118.

Srivastava, A., Babu, A., Filant, J., Moxley, K.M., Ruskin, R., Dhanasekaran, D., Sood, A.K.,

McMeekin, S., Ramesh, R., 2016. Exploitation of exosomes as nanocarriers for gene-, chemo-,

and immune-therapy of cancer. J. Biomed. Nanotechnol. 12, 1159-1173.

ACCEPTED MANUSCRIPT

Page 33: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

32

Sterzenbach, U., Putz, U., Low, L.H., Silke, J., Tan, S.S., Howitt, J., 2017. Engineered exosomes

as vehicles for biologically active proteins. Mol. Ther. 25, 1269-1278.

Stranford, D.M., Leonard, J.N., 2017. Delivery of biomolecules via extracellular vesicles: A

budding therapeutic strategy, in: Friedmann, T., Dunlap, J.C., Goodwin, S.F. (Eds.), Adv. Genet.

98, 155-175.

Stremersch, S., Vandenbroucke, R.E., Van Wonterghem, E., Hendrix, A., De Smedt, S.C.,

Raemdonck, K., 2016. Comparing exosome-like vesicles with liposomes for the functional

cellular delivery of small RNAs. J. Control. Release 232, 51-61.

Sugahara, K.N., Teesalu, T., Karmali, P.P., Kotamraju, V.R., Agemy, L., Girard, O.M., Hanahan,

D., Mattrey, R.F., Ruoslahti, E., 2009. Tissue-penetrating delivery of compounds and nanoparticles

into tumors. Cancer Cell 16, 510-520.

Sullivan, R., Maresh, G., Zhang, X., Salomon, C., Hooper, J., Margolin, D., Li, L., 2017. The

emerging roles of extracellular vesicles as communication vehicles within the tumor

microenvironment and beyond. Front. Endocrinol. (Lausanne) 8, 194.

Sun, D.M., Zhuang, X.Y., Xiang, X.Y., Liu, Y.L., Zhang, S.Y., Liu, C.R., Barnes, S., Grizzle, W.,

Miller, D., Zhang, H.G., 2010. A novel nanoparticle drug delivery system: The anti-inflammatory

activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 18, 1606-1614.

Takahashi, Y., Nishikawa, M., Shinotsuka, H., Matsui, Y., Ohara, S., Imai, T., Takakura, Y., 2013.

Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice

after intravenous injection. J. Biotechnol. 165, 77-84.

Tamura, R., Uemoto, S., Tabata, Y., 2017. Augmented liver targeting of exosomes by surface

modification with cationized pullulan. Acta Biomater. 57, 274-284.

Thery, C., Ostrowski, M., Segura, E., 2009. Membrane vesicles as conveyors of immune

responses. Nat. Rev. Immunol. 9, 581-593.

Thery, C., Zitvogel, L., Amigorena, S., 2002. Exosomes: Composition, biogenesis and function.

Nat. Rev. Immunol. 2, 569-579.

Tian, T., Wang, Y., Wang, H., Zhu, Z., Xiao, Z., 2010. Visualizing of the cellular uptake and

intracellular trafficking of exosomes by live-cell microscopy. J. Cell. Biochem. 111, 488-496.

Tian, T., Zhang, H.-X., He, C.-P., Fan, S., Zhu, Y.-L., Qi, C., Huang, N.-P., Xiao, Z.-D., Lu, Z.-H.,

Tannous, B.A., Gao, J., 2018. Surface functionalized exosomes as targeted drug delivery vehicles

for cerebral ischemia therapy. Biomaterials 150, 137-149.

Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G.J., Wei, J., Nie, G., 2014. A doxorubicin

delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy.

Biomaterials 35, 2383-2390.

Tiwari, P.M., Eroglu, E., Bawage, S.S., Vig, K., Miller, M.E., Pillai, S., Dennis, V.A., Singh, S.R.,

2014. Enhanced intracellular translocation and biodistribution of gold nanoparticles functionalized

with a cell-penetrating peptide (VG-21) from vesicular stomatitis virus. Biomaterials 35,

9484-9494.

Tkach, M., Thery, C., 2016. Communication by extracellular vesicles: Where we are and where

we need to go. Cell 164, 1226-1232.

Valadi, H., Ekstrom, K., Bossios, A., Sjostrand, M., Lee, J.J., Lotvall, J.O., 2007.

Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange

between cells. Nat. Cell Biol. 9, 654-U672.

van Asbeck, A.H., Beyerle, A., McNeill, H., Bovee-Geurts, P.H.M., Lindberg, S., Verdurmen,

ACCEPTED MANUSCRIPT

Page 34: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

33

W.P.R., Hallbrink, M., Lange, U., Heidenreich, O., Brock, R., 2013. Molecular parameters of

siRNA-cell penetrating peptide nanocomplexes for efficient cellular delivery. Acs Nano 7,

3797-3807.

Van den Boorn, J.G., Schlee, M., Coch, C., Hartmann, G., 2011. SiRNA delivery with exosome

nanoparticles. Nat. Biotechnol. 29, 325-326.

van der Meel, R., Fens, M.H.A.M., Vader, P., van Solinge, W.W., Eniola-Adefeso, O., Schiffelers,

R.M., 2014. Extracellular vesicles as drug delivery systems: Lessons from the liposome field. J.

Control. Release 195, 72-85.

van Dommelen, S.M., Vader, P., Lakhal, S., Kooijmans, S.A.A., van Solinge, W.W., Wood, M.J.A.,

Schiffelers, R.M., 2012. Microvesicles and exosomes: Opportunities for cell-derived membrane

vesicles in drug delivery. J. Control. Release 161, 635-644.

Vlassov, A.V., Magdaleno, S., Setterquist, R., Conrad, R., 2012. Exosomes: Current knowledge of

their composition, biological functions, and diagnostic and therapeutic potentials. Biochim.

Biophys. Acta-Gen. Subjects 1820, 940-948.

Wan, Y., Wang, L., Zhu, C., Zheng, Q., Wang, G., Tong, J., Fang, Y., Xia, Y., Cheng, G., He, X.,

Zheng, S.-Y., 2018. Aptamer-conjugated extracellular nanovesicles for targeted drug delivery.

Cancer Res. 78, 798-808.

Wang, C.-F., Makila, E.M., Kaasalainen, M.H., Liu, D., Sarparanta, M.P., Airaksinen, A.J.,

Salonen, J.J., Hirvonen, J.T., Santos, H.A., 2014. Copper-free azide-alkyne cycloaddition of

targeting peptides to porous silicon nanoparticles for intracellular drug uptake. Biomaterials 35,

1257-1266.

Watson, D.C., Bayik, D., Srivatsan, A., Bergamaschi, C., Valentin, A., Niu, G., Bear, J., Monninger,

M., Sun, M., Morales-Kastresana, A., Jones, J.C., Felber, B.K., Chen, X., Gursel, I., Pavlakis, G.N.,

2016. Efficient production and enhanced tumor delivery of engineered extracellular vesicles.

Biomaterials 105, 195-205.

Wiklander, O.P.B., Nordin, J.Z., O'Loughlin, A., Gustafsson, Y., Corso, G., Mäger, I., Vader, P., Yi,

L., Sork, H., Seow, Y., 2015. Extracellular vesicle in vivo biodistribution is determined by cell

source, route of administration and targeting. J. Extracellular Vesicles 4, 26316.

Yanez-Mo, M., Siljander, P.R.M., Andreu, Z., Zavec, A.B., Borras, F.E., Buzas, E.I., Buzas, K.,

Casal, E., Cappello, F., Carvalho, J., Colas, E., Cordeiro-da Silva, A., Fais, S., Falcon-Perez, J.M.,

Ghobrial, I.M., Giebel, B., Gimona, M., Graner, M., Gursel, I., Gursel, M., Heegaard, N.H.H.,

Hendrix, A., Kierulf, P., Kokubun, K., Kosanovic, M., Kralj-Iglic, V., Kramer-Albers, E.-M.,

Laitinen, S., Lasser, C., Lener, T., Ligeti, E., Line, A., Lipps, G., Llorente, A., Lotvall, J.,

Mancek-Keber, M., Marcilla, A., Mittelbrunn, M., Nazarenko, I., Nolte-'t Hoen, E.N.M., Nyman,

T.A., O'Driscoll, L., Olivan, M., Oliveira, C., Pallinger, E., Del Portillo, H.A., Reventos, J., Rigau,

M., Rohde, E., Sammar, M., Sanchez-Madrid, F., Santarem, N., Schallmoser, K., Ostenfeld, M.S.,

Stoorvogel, W., Stukelj, R., Van der Grein, S.G., Vasconcelos, M.H., Wauben, M.H.M., De Wever,

O., 2015. Biological properties of extracellular vesicles and their physiological functions. J.

Extracellular Vesicles 4, 27066.

Yang, J., Zhang, X., Chen, X., Wang, L., Yang, G., 2017a. Exosome mediated delivery of miR-124

promotes neurogenesis after ischemia. Mol. Ther.-Nucl. Acids 7, 278-287.

Yang, T., Fogarty, B., LaForge, B., Aziz, S., Thuy, P., Lai, L., Bai, S., 2017b. Delivery of Small

Interfering RNA to Inhibit Vascular Endothelial Growth Factor in Zebrafish Using Natural Brain

Endothelia Cell-Secreted Exosome Nanovesicles for the Treatment of Brain Cancer. AAPS J. 19,

ACCEPTED MANUSCRIPT

Page 35: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

34

475-486.

Yang, T., Martin, P., Fogarty, B., Brown, A., Schurman, K., Phipps, R., Yin, V.P., Lockman, P., Bai,

S., 2015. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer

rherapy in Danio Rerio. Pharm. Res. 32, 2003-2014.

Yang, Y.S., Hong, Y.S., Nam, G.H., Chung, J.H., Koh, E., Kim, I.S., 2017c. Virus-mimetic

fusogenic exosomes for direct delivery of integral membrane proteins to target cell membranes.

Adv. Mater. 29, 1605604.

Yuan, D.F., Zhao, Y.L., Banks, W.A., Bullock, K.M., Haney, M., Batrakova, E., Kabanov, A.V.,

2017. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain.

Biomaterials 142, 1-12.

Zhang, W., Yu, Z.L., Wu, M., Ren, J.G., Xia, H.F., Sa, G.L., Zhu, J.Y., Pang, D.W., Zhao, Y.F.,

Chen, G., 2017. Magnetic and folate functionalization enables rapid isolation and enhanced

tumor-targeting of cell-derived microvesicles. Acs Nano 11.

Zhao, C., Busch, D.J., Vershel, C.P., Stachowiak, J.C., 2016. Multifunctional Transmembrane

Protein Ligands for Cell-Specific Targeting of Plasma Membrane-Derived Vesicles. Small 12,

3837-3848.

ACCEPTED MANUSCRIPT

Page 36: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

35

Figure and Table captions:

Fig. 1. Schematic illustration of EV modification techniques. Functionalized EVs can be

generated by using “Cell engineering”, wherein EV producing cells are genetically manipulated.

Alternatively, EVs can be directly functionalized through “EV engineering”.

Fig. 2. Schematic representation of functional ligands with divergent functions that can be

introduced to EVs for improved delivery properties. EVs can be endowed with prolonged

circulation time, increased targeting capacity, and enhanced cytoplasmic delivery efficiency by

incorporation of PEG moieties (e.g., VEGF nanobody-PEG and AA-PEG), targeting ligands (e.g.,

targeting peptides, nanobodies, and SIRPα), and functional proteins or peptides (e.g., VSV-G,

GALA, and CPPs).

Fig. 3. Schematic depiction of functionalized EVs to achieve efficient cytoplasmic delivery of

their payloads. Decoration of EVs in PEG corona (e.g., EGFR-nanobody-PEG) enables them to

escape from rapid phagocytosis by the MPS and permit long circulation half-life. Modification of

EVs with targeting ligands (e.g., GE11) allows them to bind target cells via receptor-ligand

interaction. Furthermore, EVs equipped with viral proteins (e.g., VSV-G) allow for direct

cytoplasmic delivery of their payloads via membrane fusion. Arginine-rich CPP-modifed EVs can

actively induce macropinocytosis for efficient cellular uptake. After cellular uptake via

receptor-mediated endocytosis or macropinocytosis, EVs containing fusogenic ligands (e.g.

GALA) can efficiently escape from endosome for cargo release to the cytoplasm of recipient cell

via backfusion with endosome membrane.

Table 1. Overview of advantages and disadvantages of EV modification techniques.

Table 2. Ligands used to endow EVs with prolonged circulation time.

Table 3. Ligands used to equip EVs with increased targeting capacity.

Table 4. Ligands used to arm EVs with enhanced cytoplasmic delivery efficiency.

ACCEPTED MANUSCRIPT

Page 37: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

36

Table 1

Overview of advantages and disadvantages of EV modification techniques.

Modification

strategies

Advantages Disadvantages Ref.

Cell engineering

Fusion with EV

membrane

proteins

Incorporation of EVs with

preserved direction and

function

Inefficient, complex, and

time-consuming;

Variable performance;

Possible degradation of

fused ligands during EV

biogenesis;

Possibly compromise the

functionality of EVs’

membrane proteins;

May not be readily

applicable to all ligands

(Zhao et al.,

2016)

(Alvarez-Erviti

et al., 2011)

(Hung and

Leonard, 2015)

(Armstrong et

al., 2017)

Fusion with GPI No impairment of

structural and functional

integrity of EVs’

membrane proteins;

Enrichment of functional

ligands compared with

parent cells;

Applicable to multiple

ligands

Inefficient, complex, and

time-consuming protocol;

Possible degradation of

fused ligands in the

endosome during EV

biogenesis;

May not be readily

applicable to all ligands

(Kooijmans et

al., 2016a)

(Kim and

Eberwine,

2010)

Liposome-based

cellular

engineering

Efficient and controlled

protocols;

Without modification of

EVs membrane proteins;

Allows for simultaneous

incorporation of EVs with

multiple functional

ligands;

Possibly applicable to

multiple ligands

Possibly complex and

time-consuming

(Lee et al.,

2016)

Mechanical

extrusion

Rapid and easy to

perform;

Economic production

process

Likely to alter the structure

and orientation of ligands

and EVs’ membrane proteins

during the extrusion process

Unknown immunogenicity

(Wan et al.,

2018)

Packaging

exogenous

No modification of EVs’

membrane proteins;

Difficult to target deep

tissues in vivo;

(Silva et al.,

2013)

ACCEPTED MANUSCRIPT

Page 38: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

37

materials Applicable to a wide array

of EVs and ligands

Potential toxic issues of

exogenous materials

(Silva et al.,

2015)

EV engineering

Chemical

conjugation

Rapid and easy to

perform;

Controllable and scalable

protocols;

Compatible with some

chemical reagents and

reaction conditions that

can not be used in “Cell

engineering”;

Have general applicability

to a wide range of ligands;

Allow for incorporation of

high ligand density

Excessive conditions may

impair membrane integrity

and denature the

functionality of EVs’

membrane proteins;

Induce potential

immunogenicity and toxicity;

Requirement of complex

purification steps

(Smyth et al.,

2014)

(Tian et al.,

2018)

Post insertion Fast and simple

manipulation;

Mild modification

condition;

Have general applicability

to multiple EVs and ligand

types

Likely low efficiency of ligand

incorporation;

Unknown ligand

incorporation stability

(Kooijmans et

al., 2016b)

(Kim et al.,

2018)

Electrostatic

interaction

Fast and simple

manipulation;

Mild modification

condition;

Have general applicability

to multiple EVs and ligand

types

Possibly low efficiency of

ligand incorporation;

Unknown ligand

incorporation stability

(Tamura et al.,

2017)

ACCEPTED MANUSCRIPT

Page 39: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

38

Table 2

Ligands used to endow EVs with prolonged circulation time.

Ligands Modification

approach

Origin of EVs Outcome Ref.

EGFR-nanobody-PEG Post insertion Human

epidermoid

carcinoma cells

Prolonged

circulation

time and

improved cell

specificity

(Kooijmans et

al., 2016b)

AA-PEG Post insertion Macrophages Prolonged

circulation

time and

improved cell

specificity

(Kim et al.,

2018)

ACCEPTED MANUSCRIPT

Page 40: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

39

Table 3

Ligands used to equip EVs with increased targeting capacity.

Targeting ligands Modificati

on

approach

Origin of

EVs

Target Outcome Ref.

RVG peptide Fusion

with

Lamp2b

imDCs Acetylcholine

receptor

Greater

accumulation

in the brain

and specific

gene

knockdown

(Alvarez-Er

viti et al.,

2011)

(Liu et al.,

2015)

iRGD peptide Fusion

with

Lamp2b

imDCs αν-integrin Significant

inhibition of

breast cancer

without overt

toxicity

(Tian et al.,

2014)

GE11 peptide Fusion

with

PDGFR

HEK293

cells

EGFR Marked

suppression of

breast cancer

in mice

(Ohno et

al., 2013)

IL3 peptide Fusion

with

Lamp2b

HEK293T

cells

IL3 receptors

overexpressed

on CML

Significant

regression of

cancer cell

growth

(Bellavia

et al.,

2017)

c(RGDyK) Click

chemistry

MSCs integrin αvβ3 Efficiently

suppression of

inflammarory

response in

lesion region

(Tian et al.,

2018)

EGFR nanobody Fusion

with GPI

Neuro2A

cells

EGFR

overexpressed

tumor cells

Increased

tumor

targeting

capacity

(Kooijmans

et al.,

2016a)

EGFR-nanobody-PE

G

Post

insertion

Neuro2A

cells

EGFR

overexpressed

tumor cells

Improved

targeting

capacity and

extended

circulation

time

(Kooijmans

et al.,

2016b)

SIRPα Fusion

with

PDGFR

HEK293T

cells

CD47 Remarkably

augmented

tumor

phagocytosis

(Koh et al.,

2017)

ACCEPTED MANUSCRIPT

Page 41: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

40

and anti-tumor

T cell

responses

Lipid-conjugated

AS1411 aptamer

Hydropho

bic

insertion

DCs Necleolin Significantly

improved

targeting

capacity and

therapeutic

efficacy

(Wan et al.,

2018)

Cholesterol-conjug

ated EGFR aptamer

Post

insertion

HEK293T

cells

EGFR Significant

inhibition of

orthotopic

breast cancer

(Pi et al.,

2018)

FA Cell

engineerin

g

Bovine

milk;

Macrophag

es

FA receptor Improved

antitumor

efficacy

(Munagala

et al.,

2016)

(Zhang et

al., 2017)

Cationized pullulan Electrostat

ic

interaction

MSCs Hepatocyte

asialoglycoprot

ein receptors

Enhanced

anti-inflammat

ory effect in

liver injury

(Tamura et

al., 2017)

AA-PEG Post

insertion

Macrophag

es

Sigma receptor Greater

targeting

capacity and

extended

circulation

time

(Kim et al.,

2018)

Iron oxide

nanoparticles

Cell

engineerin

g

Macrophag

es

- Enhanced

anti-tumor

effect

(Silva et al.,

2015 )

SMCNCs Cell

engineerin

g

Blood - Increased

anti-tumor

effect

(Qi et al.,

2016)

ACCEPTED MANUSCRIPT

Page 42: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

ACC

EPTE

D M

ANU

SCR

IPT

41

Table 4

Ligands used to arm EVs with enhanced cytoplasmic delivery efficiency.

Ligands Modification

approach

Origin of EVs Outcome Ref.

VSV-G protein Cell engineering HEK293T cells Mediating membrane

fusion for cargo delivery

directing recipient cell

membrane

(Yang et al.,

2017c )

GALA peptide

and cationic

lipids

EV engineering;

electrostatic

interaction

Hela cells Efficient cellular uptake

and cytoplasmic release

(Nakase et

al., 2015)

GALA peptide Cell

engineering;

chemical

conjugation

Murine

melanoma

B16BL6 tumor

cells

Efficient cytosolic delivery

and enhanced tumor

antigen presentation

(Morishita

et al., 2017)

Arginine-rich

CPPs (R8)

Chemical

conjugation

Hela cells Activation of

macropinocytosis and

increased cellular uptake

(Nakase et

al., 2016)

Arginine-rich

CPPs (R4, R8,

R12, R16)

Chemical

conjugation

Hela cells R16 CPPs achieved

maximum cellular uptake

and showed effective

anticancer activity

(Nakase et

al., 2017)

L17E peptide Post insertion Hela cells Perturbation of endosomal

membrane and efficient

cytoplasmic delivery

(Akishiba et

al., 2017)

ACCEPTED MANUSCRIPT

Page 43: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

Graphics Abstract

Page 44: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

Figure 1

Page 45: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

Figure 2

Page 46: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

Figure 3

Page 47: Functionalized extracellular vesicles as advanced ...download.xuebalib.com/2ph7JZ9r5yfB.pdf · ACCEPTED MANUSCRIPT 1 Functionalized extracellular vesicles as advanced therapeutic

本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP

图书馆。

图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具