You are on page 1of 10

BASIC SCIENCE

Nanomedicine: Nanotechnology, Biology, and Medicine


14 (2018) 195 – 204

Original Article nanomedjournal.com

Engineering macrophage-derived exosomes for targeted paclitaxel


delivery to pulmonary metastases: in vitro and in vivo evaluations
Myung Soo Kim, PhD a, b , Matthew J. Haney a, b , Yuling Zhao a, b , Dongfen Yuan a, b ,
Irina Deygen, PhD c , Natalia L. Klyachko, PhD a, b, c , Alexander V. Kabanov, PhD a, b, c ,
Elena V. Batrakova, PhD a, b,⁎
a
Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
b
Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
c
Deparment of Chemical Enzymology, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia
Received 20 April 2017; accepted 19 September 2017

Abstract

Exosomes have recently emerged as a promising drug delivery system with low immunogenicity, high biocompatibility, and high efficacy
of delivery. We demonstrated earlier that macrophage-derived exosomes (exo) loaded with a potent anticancer agent paclitaxel (PTX)
represent a novel nanoformulation (exoPTX) that shows high anticancer efficacy in a mouse model of pulmonary metastases. We now report
the manufacture of targeted exosome-based formulations with superior structure and therapeutic indices for systemic administration. Herein,
we developed and optimized a formulation of PTX-loaded exosomes with incorporated aminoethylanisamide-polyethylene glycol (AA-PEG)
vector moiety to target the sigma receptor, which is overexpressed by lung cancer cells. The AA-PEG-vectorized exosomes loaded with PTX
(AA-PEG-exoPTX) possessed a high loading capacity, profound ability to accumulate in cancer cells upon systemic administration, and
improved therapeutic outcomes. The combination of targeting ability with the biocompatibility of exosome-based drug formulations offers a
powerful and novel delivery platform for anticancer therapy.
Published by Elsevier Inc.

Key words: Drug delivery systems; Exosomes; Paclitaxel; Pulmonary metastases

The lung is one of the most common sites of metastases and poor; chemotherapy provides only minimal increases in survival
tumor relapse following treatment of the primary tumor mass; rates, with a five-year survival rate of less than 15%. 2,3 Thus, the
lung metastases are identified in 30%-55% of all cancer patients. efficient, targeted delivery of anticancer agents to pulmonary
Because pulmonary metastases are distributed throughout the metastases remains one of the greatest challenges for the therapy.
pulmonary parenchyma, their excision is difficult, if not The recently emerged field of nanotechnology holds great
impossible. Clinical reports have highlighted a rare occurrence promise for developing drug delivery systems with targeting and
of complete regression for metastatic tumors in patients who controlled-release characteristics for cancer treatment; there have
underwent standard-of-care palliative treatment involving radi- been many new advances and innovations made in this field
ation or immunotherapy. As a result, lung cancer is the leading during the past decade. 1 A large proportion of chemotherapeutic
cause of cancer-related deaths in the world, responsible for more drugs have low aqueous solubility, consequently requiring the
deaths than breast, colon, and prostate cancers combined. 1 In use of specialized delivery vehicles (e.g. micelles, liposomes,
particular, non-small cell lung cancer (NSCLC) is responsible for polymeric nanoparticles, or other types of nanoparticles) for
~85% of all lung cancers, and prognosis of metastatic NSCLC is parenteral administration. However, these nanosized delivery

This study was supported by the United States National Institutes of Health grants 1RO1 NS057748 and The Carolina Partnership, a strategic partnership
between the UNC Eshelman School of Pharmacy and The University Cancer Research Fund through the Lineberger Comprehensive Cancer Center (to EVB),
RR021937 (to AVK), and grant RSF-14-13-00731 (to both AVK and NLK).
⁎Corresponding author at: UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7362.
E-mail address: batrakov@ad.unc.edu (E.V. Batrakova).

https://doi.org/10.1016/j.nano.2017.09.011
1549-9634/Published by Elsevier Inc.

Please cite this article as: Kim MS, et al, Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and
in vivo evaluations. Nanomedicine: NBM 2018;14:195-204, https://doi.org/10.1016/j.nano.2017.09.011
196 M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204

vehicles are often difficult to manufacture and may cause Based on these findings, we hypothesized that modification
unwanted side effects (such as the excipient Cremophor EL in of PTX-loaded exosomes with PEG-AA vector moiety will
the commercial formulation of PTX, Taxol). In addition, improve their circulation time in the blood and allow to target
conventional drug nanoformulations normally are cleared pulmonary metastases. Our investigations revealed a robust
rapidly from the circulation by the mononuclear phagocyte accumulation and nearly complete co-localization of systemi-
system (MPS). 4 This demands innovative approaches for cally administered AA-exosomes with pulmonary metastases,
engineering drug delivery systems. and greater in vivo therapeutic efficacy as compared to
Exosomes are membrane-derived vesicles ~40-200 nm in non-vectorized exoPTX and Taxol. Thus, exosome-based
diameter 5; they may be found in extracellular bodily fluids and in formulations may represent the next generation of drug delivery
conditioned cell culture media. 6 Exosomes are released by a systems that combines nanoparticle size with targeted drug
variety of cell types; and formed when multi-vesicular bodies delivery and low immunogenic profile.
inside the cells fuse with the plasma membrane and release
intraluminal vesicles into the extracellular microenvironment. 6,7
Exosomes naturally function as intracellular messengers, Materials and methods
carrying RNAs and proteins between the cells. 8 Recently,
exosomes have begun to be explored for use as drug delivery Description of reagents, cell culture conditions, animals used
vehicles for non-native therapeutics such as nucleic acids, 9–14 in these studies, characterization of exosomal formulations as
therapeutic proteins, 15 and small molecule drugs. 6,16,17 The well as accumulation and intracellular trafficking experiments
membranotropic nature of exosomes suggests that these drug and statistical analysis are described in Supplementary Material.
carriers will be able efficiently interact with the target cancer
cells and deliver their toxic payload. This process is facilitated by Preparation of AA-vectorized exosomes targeted to the sigma
membrane interactions and fusion due to the expression of receptor
various adhesive proteins (tetraspanins and integrins) on the
For all in vitro experiments, exosomes were harvested from
surface of exosomes. 18 Noteworthy, it was reported that
the supernatants of RAW 264.7 cells cultured in
exosomes are able to deliver their intraluminal cargo into the
exosome-depleted media using the ExoQuick-TC™ Kit as
cytosol of target cells. 19 In addition, allogenic exosomes have an
described earlier. 22 For all in vivo experiments, exosomes
immune privileged status, which allows for decreased drug
released by primary bone-marrow derived macrophages (BMM)
clearance by MPS. Specifically, immunocytes-derived exosomes
isolated from C57BL/6 mice were utilized. Exosomes targeted to
are known to express CD47 receptor, 20 which interacts with
sigma receptor with DSPE-PEG-AA (AA-PEG-exo) and
signal regulatory protein α (SIRPα) to produce a “don't eat me”
non-vectorized control exosomes with DSPE-PEG (PEG-exo)
signal in phagocytes. 21 These unique features make exosomes
were prepared as follows: exosomes were isolated from
an attractive option for use as a drug delivery vehicle for
macrophage media as described above and then DSPE-PEG or
cancer treatment.
DSPE-PEG-AA (50 μg/ml) was added to the mixture (for
We reported earlier that macrophage-derived exosomes can
PEG-exo and AA-PEGexo, respectively). 100 μL PTX (10 mg/mL
be loaded with low molecular chemotherapeutics, such as PTX,
in ethanol) was also added to the exosomes to incorporate PTX
and doxorubicin (DOX), 22 or therapeutic proteins, such as
using sonication method that was developed in our laboratory. 22
catalase 15 or brain derived neurotrophic factor, BDNF. 23
After sonication, AA-PEGexo, or PEGexo, or AA-exoPTX
Different loading procedures (co-incubation with a drug at
solutions were incubated at 37 °C for 60 min to allow for recovery
room temperature, electroporation, saponin permeabilization,
of the exosomal membrane. Exosomes were purified from the
extrusion, freeze–thaw cycles, or sonication) were utilized to
excess of free DSPE-PEG or DSPE-PEG-AA by size exclusion
incorporate these therapeutic agents into exosomes. We
chromatography using a NAP-10 Sephadex G25 column (GE
demonstrated that a reformation of exosomal membranes upon
Healthcare, Buckinghamshire, UK). Purified exosomal formula-
sonication allowed efficient drug loading along with preservation
tions were collected and stored at −20 °C.
of exosomal structure and internal content. Here we report the
development of improved exosome-based PTX formulation Drug loading and optimization AA-PEG-exoPTX
targeted to cancer cells for systemic administration. It has been
shown that a variety of cancer types, including NSCLC, For PTX loading into vectorized exosomes, 1 mL of purified
overexpress sigma receptor, 24 a membrane-bound protein with exosomes (~10 11 exosomes) in PBS was mixed with PTX and
an as-yet undefined role. AA is a ligand with high affinity for DSPE-PEG-AA. For this purpose, first PTX (10 mg/mL drug in
sigma receptor and has been utilized to targeted delivery of Dox, EtOH stock solution) was added to 1 mL exosomes in PBS.
proteins, and siRNA. 25–27 Next, the most common method of Then, different amounts of AA-PEG-DSPE (0.05-0.50 mg/ml) in
reducing the immunogenicity of nanoformulated drugs is to PBS were added to the mixture of exosome with PTX. The
decorate the nanoparticle in a polyethylene glycol (PEG) corona, obtained mixture was sonicated using a Model 505 Sonic
which reduces recognition by the MPS and aids in avoiding Dismembrator using .25″ tip (Thermo Fisher Scientific, USA) as
clearance. To this end, Kooijmans et al have recently shown that described earlier. 22 After the sonication, a solution
the introduction of polyethylene glycol to exosomes results in AA-vectorized exosomes loaded with PTX (AA-PEG-exoPTX)
stealth properties, which significantly increases their circulation were incubated at 37 °C for 60 min to allow for recovery of the
time in mice. 28 exosomal membrane. The excess free PTX and AA-PEG-DSPE
M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204 197

were eliminated from AA-PEG-exoPTX by size exclusion 4, and 7; 0.5 mg/kg totally), or Taxol (same regiment as
chromatography as described elsewhere. 22 The amount of PTX exosome-based formulations), or saline as a control (n = 7). To
loaded into exosomes was measured by HPLC method. 22 All assess amount of cancer metastases, two mice from each group
analyses were performed using a C18 column (Supelco Nucleosil were sacrificed on day 18, perfused, and lung slides were
C18, 250 mm × 4.6 mm, 5 μm, 100 Å, Sigma-Aldrich,) with a examined by confocal microscopy. The rest of the mice (n = 5)
mobile phase of H2O:acetonitrile (45:55, v/v) at a flow rate of were monitored daily for the signs of the reduced physical
1 mL/min at 30 °C. Loading capacity is expressed by μg protein activity and the progression of the tumor. The survive time of
of exosomes. each mouse was recorded. To perform quantitative analysis, an
additional experiment with the same treatment groups of mice
Biodistribution of intravenously injected exosomes in mice with (n = 5) was carried out. On day 18 mice were sacrificed,
pulmonary metastases perfused, and lung slides were examined by confocal microsco-
py. The metastases area on the slides was calculated using
To utilize fluorescence imaging, 3LL-M27 cells were trans-
ImageJ software. The comparison was performed in randomized
duced with lentiviral vectors encoding the optical reporter FITC
order of the blinded experiment on 10-15 sets of images acquired
(FITCFlmC, green) fluorescent protein as reported earlier. 29 The
with the same optical settings.
viral construct also encoded for a puromycin resistance gene
downstream of FITC, which was introduced to enable for the
Statistical analysis
selection of positively transduced cells. C57BL/6 mice were
injected intra tail vein (i.v.) with FITC-FLmC-3LL-M27 cells For the all experiments, data are presented as the mean ±
(5 × 10 6 cells/mouse in 100 μl saline) and pulmonary metastases S.E.M. Tests for significant differences between the groups
were allowed to establish. In parallel, exosomes isolated from were performed using a t-test or one-way ANOVA with multiple
autologous macrophages conditioned media were stained with DiL comparisons (Fisher's pairwise comparisons) using GraphPad
(red) and vectorized to sigma receptor with DSPE-PEG-AA. To Prism 5.0 (GraphPad software, San Diego, CA, USA). A
stain exosomes with DiL, exosome pellet was rehydrated in 500 μL minimum P value of 0.05 was chosen as the significance level.
PBS, supplemented with 1 mM stock solution DiL in DMSO
(5 μL), and incubated for 20 minutes at RT. Following incubation,
exosomes were isolated by PEG precipitation and rehydrated in Results
500 μL PBS. The fluorescently-labeled exosomes were addition-
ally purified from remaining non-incorporated DiL on NAP 10 Manufacture and characterization of AA-PEG-exoPTX
column. Twelve days following cancer cells injection, mice with
We demonstrated earlier that efficient loading of PTX can be
established lung metastases were intravenously (i.v.) injected with
achieved, when exosomes are subjected to ultrasound treatment
autologous vectorized exosomes DiL-AA-PEG-exo, or
in the presence of the drug. 22 Herein, we applied the same
non-vectorized exosomes DiL-exo as a control group (10 8
approach for simultaneous loading of PTX, and vectorization of
particles/100 μl, n = 4 per group). Four hours later, mice were
exosomes to sigma receptor using AA moiety. Obtained
sacrificed, perfused, lungs were extracted and sectioned at a
formulations were characterized by size, charge, protein content,
thickness of 20 μm; nuclei were stained with DAPI (300 mM, 5
and PTX loading capacity (LC). First, the effect of incorporation
min). The images of lung and liver sections were examined by a
of vector lipid molecule (DSPE-PEG-AA) on the LC was
confocal fluorescence microscopic system ACAS-570 (Meridian
evaluated. LC was expressed as the amount of the drug vs. the
Instruments, Okimos, MI) with argon ion laser and corresponding
amount of exosomal protein. HPLC analysis revealed that
filter set. Digital images were obtained using the CCD camera
incorporation of high amounts of AA-PEG-DSPE (0.5 mg/ml) into
(Photometrics) and Adobe Photoshop software. Quantification of
exosomes significantly decreased their LC for PTX (Figure 1, A). In
immunostaining was performed with ImageJ software, utilizing
contrast, lower amounts of the lipid (0.25-0.05 mg/ml) did not
JACoP plugins to calculate Pearson's co-localization
affect LC for PTX. It is likely that excess of hydrophobic chains
coefficients. 30 The comparison was performed in randomized
of the lipid incorporated into exosomal membranes diminished
order of the blinded experiment on 10-15 sets of images acquired
available for PTX space. Therefore, we choose the high amount
with the same optical settings.
of the lipid (0.25 mg/ml) that did not significantly reduce PTX
Therapeutic efficacy of AA-PEG-exoPTX against pulmonary loading to vectorize exosomes. LC for the optimal
metastases AA-PEG-exoPTX formulation was ~33%, comparable to the
LC achieved for non-vectorized exoPTX (Figure 1, A).
The antineoplastic effects of PTX exosome formulation were Noteworthy, ultrasound treatment significantly increased
evaluated in a mouse model of pulmonary metastases with amount of PTX incorporated into exosomes; the LC in
3LL-M27 cells transduced with lentiviral vectors encoding the exosomes without sonication was as low as 1.4%. 22
optical reporter mCherry (GBM8FlmC, red) as described To address a concern about possible alterations in the exosomal
earlier. 22 To establish pulmonary metastases, C57BL/6 mice membranes upon incorporation of vector moiety (AA-PEG-DSPE)
were i.v. injected with 8FlmC-3LL-M27 cancer cells (5 × 10 6 using sonication, the levels of exosome-specific proteins, TSG101
cells/100 μl/ mouse). Forty-eight hours later, mice were treated and flotillin, in different exosomal formulations were examined by
i.v. with autologous AA-PEG-exoPTX, or exoPTX, or empty western blot (Figure 1, B). The mild sonication utilized for PTX
exosomes (exo) (4 × 10 11 particles/100 μl, three times on day 1, loading with six cycles, and intermediate time out for cooling down
198 M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204

Figure 1. Characterization of exoPTX and AA-PEG-exoPTX formulations. PTX and vector moiety AA-PEG-DSPE were incorporated in exosomes by
sonication procedure. (A) Loading Capacity (LC) for PTX in vectorized exosomes was measured by HPLC. Increasing amounts of vector moiety resulted in LC
decrease. The highest concentration of AA-PEG-DSPE that did not lower the LC for PTX in exosomes was chosen for subsequent experiments (shown by
arrow). (B) Western blot data indicated that formulations retained the exosome markers TSG101, flotillin, and LFA1, specific marker for lymphocytes, after the
sonication procedure. (C) Size was measured by NTA and DLS; the zeta potential was measured by DLS. The data were obtained from five independent
experiments.

and membrane restoration did not affect the protein content of due to the shielding of the exosomal membrane by the long PEG
exosomes. In particular, sonicated vectorized and non-vectorized chains of the lipid (Figure 1, C).
exosomes, as well as vectorized exosomes loaded with PTX
showed elevated expression of exosome-associated proteins Effect of AA-PEG-DSPE incorporation on membrane fluidity in
(TSG101, and flotillin) as compared to cell lysate, which displayed exosomes
greater levels of β-actin (Figure 1, B). Noteworthy, we reported Fluidity of exosomal membrane upon AA-PEG-DSPE
earlier that sonication itself did not affect levels of incorporation was examined using BODIPY-PC. This is a
exosome-specific proteins compared to naïve non-sonicated hydrophobic fluorescent compound, which incorporates in the
exosomes. 22 Furthermore, exosomal formulations, as well as hydrocarbon regions of lipid membranes. Transfer of
parental macrophages, were also found to express the lymphocyte BODIPY-PC from the aqueous environment into lipid bilayers
function associated antigen-1 (LFA1, subunit CD11a) (Figure 1, B), results in a drastic increase of the fluorescence emission for this
which assists in cell uptake and may bind to endothelial cell adhesion probe. Once the probe is incorporated into lipid membranes, its
molecules overexpressed on activated endothelial cells, such as those fluorescence polarization (reflecting the ability to freely rotate in
found in tumors. 31 This is important, since the presence of LFA1 on the lipid biolayers) strongly depends on the microenvironment,
the surface may improve specific targeting of exosome-based PTX with decreases in membrane microviscosity resulting in
formulations to cancer cells. increased fluorescence polarization. In this study,
Finally, the hydrodynamic size was determined by DLS and BODIPY-PC-labeled exosomes were sonicated in the presence
NTA (Figure 1, C). Naïve empty exosomes had a narrow size of AA-PEG-DSPE lipid, and fluorescence polarization was
distribution, with an average particle diameter of 110.8 ± 4.1 nm recorded (Figure 2). Co-incubation of PTX with exosomes in the
and 75.9 ± 2.6 nm as revealed by NTA and DLS, respectively. absence of sonication did not alter membrane microviscosity;
The sonication procedure significantly increased size of however, small but statistically significant fluidization of
exosomes up to 291 ± 3.5 nm (Figure 1, C). Noteworthy, exosomal membranes was recorded when a high amount of
exosomes sonicated in the presence of PTX were smaller than lipid (0.5 μg/ml) was added to the solution. Next, significant
those sonicated without PTX. This effect may be due to the decreases (more than two times) in membrane microviscosity
stabilization of exosomal membranes by the incorporated drug. were recorded upon sonication that is consistent with our
Next, it is known that sialic acid on glycosylated lipids and previous observations. 15 The fluidity of exosomal membranes
proteins is abundant on cell membranes and contributes to the was partially restored when PTX was added to the solution.
surface charge of individual cellular membranes. In this regard, Sonication of exoPTX in the presence of the lipid further
loading of exosomes with PTX did not significantly alter the increased membrane microviscosity up to naïve non-sonicated
slightly negative change of the nanocarriers (Figure 1, C). exosomes (Figure 2). Noteworthy, the greater amount of lipid
However, vectorized AA-PEG-exoPTX formulation was found was added to the solution; the higher the microviscosity levels
to have a less negative surface charge than exoPTX, probably obtained. We hypothesize that the sonication leads to
M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204 199

accumulation of DiL-labeled vectorized exosomes (AA-PEG-exo)


was studied in target 3LL-M27 cells in vitro, and compared against
control non-vectorized sonicated exosomes (exo), as well as
exosomes with incorporated PEG-DSPE lipid without AA
targeting moiety (PEG-exo) (Figure 3, A). The obtained data
indicated that vectorized AA-PEG-exo nanocarriers were taken up
in much higher quantities than non-vectorized sonicated exo-
somes. The PEGylated exosomes without AA-targeting moiety
were taken up less than parental exosomes, probably due to the
PEG chains blocking interaction of exosomal surface proteins,
which assist in cell accumulation.
To further assess the capability of incorporated AA to target
exosomes to sigma receptor, a competitive inhibition study was
carried out in 3LL-M27 cells (Figure 3, B). In this experiment,
3LL-M27 cells were pre-treated with free AA at varying
concentrations, washed with PBS, and then equal amounts of
vectorized AA-PEG-exo along with free AA were added to the
Figure 2. Effect of AA-PEG-DSPE incorporation and PTX loading on cells for one hour. Fluorescence levels were measured; the
fluidity of exosomal membranes. BODIPY-PC-labeled exosomes were
examined by fluorescence polarization measurements. The ultrasound
amount of exosomes/μg protein was quantified and graphed
treatment significantly decreased microviscosity of exosomal membranes against the concentration of AA (Figure 3, B). Results showed a
compared to naïve exosomes. The microviscosity of sonicated exosomes was dose-dependent response of AA-PEG-exo to competitive
increased with PTX loading and then further increased with incorporation of inhibition by increasing concentrations of free AA, indicating
the lipid upon sonication. Values are means ± SEM (n = 4). Symbols indicate that AA-PEG-exo were taken up by receptor-mediated endocy-
the relative level of significance compared with naïve exosomes (P b 0.05). tosis. Noteworthy, even a large amount of free AA added to the
The data were obtained from three independent experiments. Symbols
AA-vectorized exosomes was not able completely inhibit
indicate the relative level of significance compared to protein-striped
exosomes (*P b 0.05, **P b 0.005).
exosome uptake in target cells, suggesting involvement of
other exosomal surface proteins in this process, for example
LFA1 (as demonstrated by western blot, Figure 1, B) that is
consistent with our recent report. 23
dysregulation of exosomal membranes and creation of additional The importance of exosomal surface proteins in assisting in
space for PTX molecules. This resulted in an increased LC for exosome take-up was confirmed in accumulation studies using
PTX. The incorporation of high amounts of lipid molecules upon proteinase K treatment to strip exosomes of surface proteins
sonication allowed sealing membrane bilayers that may impede (Figure 4). In this experiment, DiL-labeled sonicated
PTX loading, and as a result, diminish LC (Figure 1, A). AA-vectorized (AA-PEG-exo) and non-vectorized sonicated
exosomes (exo/sonic), as well as non-sonicated naïve exosomes
Accumulation of AA-PEG-exoPTX in target cancer cells in vitro (exo/naive) were incubated with 3LL-M27 cancer cells for
various times; and the number of taken exosomes was accessed
The ability to deliver the drug payload into target cells is by fluorescence. The obtained data indicate that accumulation
crucial for the therapeutic efficacy of exosomal formulations. levels in target cells increased in order: exo/naïve b exo/sonic b
Although the molecular function of sigma receptors is not yet AA-PEG-exo (Figure 4). This confirmed our previous reports
fully defined, there is increasing evidence that these receptors are that treatment with ultrasound improved exosome accumulation
overexpressed in many cancer cells. 24 First, we validated sigma in cancer cells, 22 as well as neuronal PC12 cells. 15 In parallel,
receptor as a target for LLC murine model of pulmonary the same exosomal formulations were treated with proteinase K,
metastases. Western blot analysis revealed high expression levels and added to the cells. The digestion of the exosomal surface
in murine LLC cells (3LL-M27), as well as murine lung proteins significantly decreased uptake by target cells in all
adenocarcinoma cells (344SQ), human small-cell lung carcino- formulations. These results clearly show the advantages of
ma cells (H69/AR), and human non-small cell lung carcinoma exosome-based drug delivery systems over common synthetic
cells (A549), and low, if any, in normal human lung fibroblasts nanocarriers related to the facilitated uptake of exosome carriers
(Hel 299) (Supplemental Figure 1, A). The protein bands were by means of surface adhesive proteins. Noteworthy, stripping of
quantified and normalized to beta actin (Supplemental Figure. 1, B). surface proteins from vectorized exosomes (AA-PEG-exo)
This indicates that the choice of AA (a sigma receptor ligand) as a decreased their transport at significantly lesser extent than
vector to target 3LL-M27 cells is appropriate for the presented non-vectorized exosomes (Figure 4), probably due to the assisted
experiments in the murine LLC model, as well as for further clinical AA-mediated accumulation in cancer cells.
evaluations. Finally, to assess the effect of AA-vectorization on
Previously, we demonstrated that accumulation levels of intracellular trafficking of exosomes, 3LL-M27 cells were
fluorescently-labeled exosomes in 3LL-M27 cells was consid- supplemented with DiL-labeled AA-PEG-exo and exo as a
erably greater (about 30 times) then accumulation of liposomes control, and then stained with MitoTracker, or LysoTracker, or
or polystyrene nanoparticles. 15 Herein, the receptor-mediated ERTracker dyes to visualize mitochondria, lysosomes, or
200 M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204

Figure 3. Accumulation of AA-vectorized exosomes in cancer cells. (A) 3LL-M27 cells were incubated with fluorescently-labeled AA-PEG-exo, or PEG-exo, or
exo for various times, and accumulation levels were measured. AA-PEG-exo were more readily taken up by 3LL-M27 as compared to exo and PEG-exo. (B)
AA-PEG-exo formulation showed a dose-dependent response to competitive inhibition by AA, indicating that this formulation entered cells by receptor
mediated endocytosis. Results are expressed as number of exosomes/μg protein vs. concentration of AA. The data were obtained from three independent
experiments.

investigated the percentage of exosomes co-localized with


cancer cells in the lungs. To induce metastases, C57BL/6 mice
were injected with GFP/3LL-M27 (5 × 10 6 cells/100 μL) as
described above. Twenty-one days later, DiL-labeled autologous
non-vectorized (exo) and AA-vectorized (AA-PEG-exo) exo-
somes were injected i.v. to C57BL/6 tumor-bearing mice. Four
hours later, mice were sacrificed, perfused; lungs were sectioned
on microtome and examined by confocal microscopy. Confocal
images revealed 94.4 ± 0.8% of AA-exosomes in immunohis-
tochemistry slides were co-localized with lung metastases
(Figure 5, D-F) indicating efficient targeting of AA-exoPTX
in vivo. In contrast, only 21.8 ± 0.2% of systemically-injected
non-vectorized exosomes were co-localized with cancer metas-
tases (Figure 5, A-C). Noteworthy, no AA-exosomes were
found in the lungs of healthy animals (Figure 5, G-J). This
suggests that systemically-administered AA-exosomes can
efficiently reach pulmonary metastases and deliver their drug
Figure 4. Importance of surface proteins on exosome accumulation in
payload to target cancer cells.
3LL-M27 cancer cells. Naïve (circle), sonicated (triangle), and AA-vector-
ized (cross) exosomes were incubated with Proteinase K to strip surface
It was reported that along with pulmonary metastases, i.v.
proteins and subsequently labeled with DiL; accumulation of Proteinase injected 3LL- M27 cells populate liver in C57BL/6 mice. 32 We
K-treated exosomes (dashed line) or control exosomes (solid line) was investigated, whether AA-vectorized exosomes were able to
examined in cancer cells. Stripping the exosomal surface proteins resulted in target liver metastases similar to pulmonary metastases. As
significant decreases in exosome accumulation levels in target cells for all expected, confocal images showed close to complete
formulations. The data were obtained from three independent experiments. co-localization of fluorescently-labeled AA-PEG-exosomes
with cancer cells in the liver (Supplemental Figure 3) suggesting
that AA-exosomal formulations may be suitable for other than
endoplasmic reticulum (ER), respectively (Supplemental Figure 2). pulmonary metastases therapy.
Confocal images revealed that exosomes preferentially distribute in
order: lysosomes N ER N mitochondria. Noteworthy, the intracel-
lular fate of exosomes was not altered by the addition of a vector to Therapeutic efficacy of AA-PEG-exoPTX against lung metastases
the sigma receptor.
C57BL/6 mice with established mCherry-3LL-M27 (red)
Co-localization of systemically-administered exosomes with metastases were systemically injected with AA-PEG-exoPTX.
pulmonary metastases in LLC mice Mice injected with exoPTX, or Taxol, or saline, or exosomes
alone (without PTX) were used in control groups. Eighteen days
To assess the ability of AA-vectorized exosomes to target later mice were sacrificed, perfused, and lung slides were
pulmonary metastases, studies were conducted in the LLC examined by confocal microscopy (Figure 6, A). The images
mouse model with 3LL-M27 cells overexpressing the optical demonstrated a superior antineoplastic efficacy of AA-exoPTX
reporter green fluorescent protein (GFP). In particular, we compared to non-vectorized exoPTX, or Taxol that resulted in
M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204 201

Figure 5. Co-localization of systemically-delivered AA-exosomes with pulmonary metastases. Exosomes were isolated from macrophages conditioned media
and labeled with DiL dye (red, A and D). C57BL/6 mice were i.v. injected with 3LL-M27 cells transduced with lentiviral vectors encoding the optical reporter
GFP fluorescent protein (green, B and E). 7 days later, tumor-bearing mice were i.v. injected with DiL-labeled non-vectorized exosomes (red, A), or
AA-exosomes (red, D). Four hours later, mice were euthanized, perfused, lungs were sectioned, and stained with DAPI (blue). Confocal images revealed
significant co-localization of AA-exosomes with metastases (94.4 + 0.8%, F) that was greater than those of non-vectorized exosomes (21.8 + 0.2%, C). No
exosomes were found in lungs of healthy animals without metastases (G-J). Bar: 20 μm. The data were obtained from three independent experiments.

the potent eradication of pulmonary metastases. Additional Discussion


images of randomly selected lung slides are shown on
Supplemental Figure 4. The quantitative assessment of metas- The efficient targeted delivery of anticancer agents to
tases area on the lung slides of animals is shown on Figure 6, B. pulmonary metastases remains one of the greatest challenges
The survival studies confirmed these results (Figure 6, C). for the treatment of lung cancer. Several nanoformulations are
Administration of AA-exoPTX formulation caused a signifi- being studied in clinical trials, or have already been approved by
cantly stronger suppression of metastases growth and greater the FDA for use in humans. 33,34 However, conventional
survival time. Noteworthy, injections of control empty (naïve) nanoparticles have limited biocompatibility, and normally are
exosomes without PTX incorporated did not slow down tumor cleared rapidly from the circulation by the MPS. 4 We have
growth in mice with pulmonary metastases. This confirms the developed a new drug delivery system that is based on natural
superior antineoplastic efficacy of AA-PEG-exoPTX upon vectors, exosomes, released by autologous macrophages for the
systemic administration, as compared to Taxol, or delivery of PTX. We utilize macrophage-derived exosomes that
non-vectorized exoPTX formulation. exert unique biological activity reflective of their origin, and
202 M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204

Figure 6. AA-PEG-ExoPTX induced potent anticancer effect against lung metastases in LLC mouse model. (A) C57BL/6 mice with established Luc/
mCherry-3LL-M27 lung metastases were i.v. treated with: saline, or Taxol, or exoPTX, or AA-PEG-exoPTX, or exosomes alone (no drug). 18 days later, mice
were sacrificed, perfused, and lungs slides were examined by confocal microscopy. AA-PEG-exoPTX treatment resulted in a potent inhibition of metastases
(red) that was more effective than treatment with Taxol or exoPTX. The bar: 50 μm. (B) Quantitative assessment of metastases area on the lung slides of animals
treated with: 1) saline, or 2) Taxol, or 3) exoPTX, or 4) AA-PEG-exoPTX, or 5) exosomes alone. **P b 0.005. (C) A survival of C57BL/6 mice with established
metastases was recorded for four treatment groups: 1) saline (dimonds), or Taxol (squares), or 3) exoPTX (triangles), or 4) AA-PEG-exoPTX (crosses), or 5)
exosomes alone (stars). A superior effect on suppression of metastases growth and greater mice survival was recorded in AA-PEG-exoPTX treatment group
(n = 6). The data were obtained from three independent experiments.

therefore, can provide advantages of both nanotechnology, and The most interesting results were obtained in the mouse LLC
cell-mediated drug delivery that is based on the innate functions model. Our data demonstrate a robust accumulation and nearly
of immune cells. complete co-localization of systemically administered
Using exosomes as drug delivery vehicles offers a number of AA-vectorized exosomes with cancer metastases. We hypothe-
benefits over common drug administration regimens; however, sized that both AA-vector, and LFA1 protein expressed on
there are some limitations and challenges that need to be exosomal membranes were responsible for this preferential
addressed. One of the major challenges is the efficient loading of accumulation in pulmonary metastases upon systemic adminis-
exosomes without significant alterations to the structure and tration. Significantly, exosomes were targeted to liver metastases
content of exosomal membranes. PTX is a highly hydrophobic as well; a complete co-localization of AA-vectorized exosomes
compound that is likely to be incorporated into the inner region with cancer cells was demonstrated in this work. Taking into
of the relatively tight and highly structured lipid bilayers of account that hydrophobic fluorescent dye DiL may serve as a
exosomes. Therefore, we developed a specific procedure model drug that has a low solubility in water (for example PTX),
wherein lipid bilayers are reshuffled upon mild sonication. To it suggests that exosomes can target cancer metastases and
target exosomal carriers to cancer cells, we incorporated a vector deliver their cytotoxic payload specifically to these cells
moiety with AA that is known to specifically bind to the sigma avoiding healthy tissues. The investigations of trafficking of
receptor using the same sonication procedure. Herein, we exosomes and their components (loaded therapeutic agents, as
optimized the amount of incorporated AA vector moiety that well as exosomal lipids, and proteins, etc.) are on the way in our
did not impede with the loading of PTX. The obtained laboratory.
AA-PEG-exoPTX formulation showed an extraordinary ability Finally, a systemic administration of AA-vectorized exoso-
to accumulate in target cancer cells; these exosomes were taken mal formulation of PTX resulted in the superior antineoplastic
up via receptor-mediated endocytosis in considerably greater effect and increased survival times in mice with pulmonary
numbers than non-vectorized exosomes in vitro. Noteworthy, metastases, as compared to exoPTX or Taxol treated animals.
addition of AA moiety did not alter intracellular trafficking of We hypothesized that this effect was due to the superior
exosomal formulations in cancer cells. therapeutic efficacy of the formulation. Specifically, the efficient
M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204 203

targeting of PTX incorporated into AA-vectorized exosomes Appendix A. Supplementary data


resulted in the potent inhibition of pulmonary metastases growth.
The confocal images and quantification of metastases area on Supplementary data to this article can be found online at
lung slides confirmed this suggestion. Furthermore, targeting of https://doi.org/10.1016/j.nano.2017.09.011.
PTX to the cancer cells may also decrease side effects of the
formulation that also will manifest in increased survival times.
Notable, LLC cells are known to express the Pgp drug efflux References
transporter in vivo. 35 We demonstrated earlier that the
incorporation of PTX into exosomes may not only increase its
1. Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics,
solubility, but also allow for overcoming of Pgp-mediated drug 2007. Clin 2007;57(1):43-66.
efflux in resistant cancer cells. This effect may be attributed to 2. Goffin J, Lacchetti C, Ellis PM, Ung YC, Evans WK. Lung Cancer
differences in internalization routes for exoPTX and Taxol. Disease Site Group of Cancer Care Ontario's Program in Evidence-
Exosomes and micelles, such as those found in Taxol, enter Based C. First-line systemic chemotherapy in the treatment of advanced
resistant cancer cells by endocytosis, however exosomes have a non-small cell lung cancer: a systematic review. J Thorac Oncol
2010;5(2):260-74.
superior accumulation due to the presence of adhesion proteins,
3. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H,
tetraspanins, integrins, immunoglobulins, proteoglycans, and et al. Gefitinib or chemotherapy for non-small-cell lung cancer with
lectins, 36 which are not found on artificial nanocarriers. mutated EGFR. Med 2010;362(25):2380-8.
Furthermore, exosomes consist of cellular membranes that may 4. Peng Q, Zhang S, Yang Q, Zhang T, Wei XQ, Jiang L, et al. Preformed
fuse with the plasma and/or endocytic membranes and deliver albumin corona, a protective coating for nanoparticles based drug
their cargo, bypassing Pgp-mediated efflux. delivery system. Biomaterials 2013;34(33):8521-30.
Moreover, it is known that exosome-mediated cell-to-cell 5. Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A,
Ratajczak MZ. Membrane-derived microvesicles: important and under-
communication is key in the battle between cancer and the
appreciated mediators of cell-to-cell communication. Leukemia
immune system. 37 Thus, Parolini et al 38 showed that exosome 2006;20(9):1487-95.
fusion with target cells occurs more efficiently under acidic 6. Kalani A, Kamat PK, Chaturvedi P, Tyagi SC, Tyagi N. Curcumin-
conditions, implying that exosomes may be taken up preferen- primed exosomes mitigate endothelial cell dysfunction during hyperho-
tially by tumors (which have an acidic microenvironment) rather mocysteinemia. Life Sci 2014;107(1–2):1-7.
than the surrounding healthy tissue. Finally, decoration of 7. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes:
exosomes with PEG chains may considerably increase their current perspectives. Proteomics 2008;8(19):4083-99.
8. Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important
circulation in the blood as was demonstrated earlier. 28
in intercellular communication. J Proteomics 2010;73(10):1907-20.
Regarding the elimination fate and blood circulation time, the 9. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery
source of exosomes is of great importance. Thus, when of siRNA to the mouse brain by systemic injection of targeted exosomes.
exosomes released by cancer cells, they efficiently accumulated Nat Biotechnol 2011;29(4):341-5.
in MPS and quickly eliminated from the blood stream. 38,39 10. Wahlgren J, De LKT, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen
Contrary, exosomes released by primary fibroblast-like mesen- P, et al. Plasma exosomes can deliver exogenous short interfering RNA
to monocytes and lymphocytes. Nucleic Acids Res 2012;40(17)e130.
chymal cells can bypass immune clearance by monocytes and
11. Pan Q, Ramakrishnaiah V, Henry S, Fouraschen S, de Ruiter PE,
macrophages that results in their prolonged blood circulation. Kwekkeboom J, et al. Hepatic cell-to-cell transmission of small silencing
The reported mechanism is related to the CD47 molecule that is RNA can extend the therapeutic reach of RNA interference (RNAi). Gut
expressed on the surface of exosomes and its binding partners 2012;61(9):1330-9.
thrombospondin-1 (TSP1) and signal regulatory protein α 12. Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV,
(SIRPα) that produce the “don't eat me” effect in Filatov MV. Exosomes are natural carriers of exogenous siRNA to
macrophages. 40 This may also enhance specificity of exosomal human cells in vitro. Cell Commun Signal 2013;11:88, https://doi.org/
10.1186/1478-811X-11-88.
formulations to cancer cells in MPS-reach organs. Further
13. Johnsen KB, Gudbergsson JM, Skov MN, Pilgaard L, Moos T, Duroux
studies of the mechanism of these superior antineoplastic effects M. A comprehensive overview of exosomes as drug delivery vehicles -
are ongoing in our lab. endogenous nanocarriers for targeted cancer therapy. Biochim Biophys
Overall, all four mechanisms mentioned here are likely to Acta 2014;1846(1):75-87.
have significant impact on AA-PEG-exoPTX anticancer activity, 14. Chen L, Charrier A, Zhou Y, Chen R, Yu B, Agarwal K, et al. Epigenetic
i.e.: (i) vector-mediated preferential accumulation in cancer cells, regulation of connective tissue growth factor by MicroRNA-214 delivery
(ii) efficient delivery of incorporated cargo into target cancer in exosomes from mouse or human hepatic stellate cells. Hepatology
2014;59(3):1118-29.
cells, (iii) bypassing Pgp-mediated drug efflux in resistant cancer
15. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al.
cells, and (iv) prolonged circulation time in the blood via Exosomes as drug delivery vehicles for Parkinson's disease therapy. J
PEGylation. In conclusion, exosomes promise an unparalleled Control Release 2015;207:18-30.
efficacy in the treatment of many life-threatening conditions, 16. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al.
including those lacking effective pharmacotherapy. Treatment of brain inflammatory diseases by delivering exosome
encapsulated anti-inflammatory drugs from the nasal region to the
brain. Mol Ther 2011;19(10):1769-79.
Acknowledgments 17. Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, et al. A novel
nanoparticle drug delivery system: the anti-inflammatory activity of
We are grateful to Dr. Leaf Huang for the invaluable curcumin is enhanced when encapsulated in exosomes. Mol Ther
comments and suggestions. 2010;18(9):1606-14.
204 M.S. Kim et al / Nanomedicine: Nanotechnology, Biology, and Medicine 14 (2018) 195–204

18. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of 30. Bolte S, Cordelieres FP. A guided tour into subcellular colocalization
extracellular vesicle uptake. J Extracell Vesicles 2014;3, https:// analysis in light microscopy. J Microsc 2006;224(Pt 3):213-32.
doi.org/10.3402/jev.v3.24641. 31. Maruo Y, Gochi A, Kaihara A, Shimamura H, Yamada T, Tanaka N, et al.
19. Montecalvo A, Larregina AT, Shufesky WJ, Stolz DB, Sullivan ML, ICAM-1 expression and the soluble ICAM-1 level for evaluating the
Karlsson JM, et al. Mechanism of transfer of functional microRNAs metastatic potential of gastric cancer. Cancer 2002;100(4):486-90.
between mouse dendritic cells via exosomes. Blood 2012;119(3):756-66. 32. Cullen R, Germanov E, Shimaoka T, Johnston B. Enhanced
20. Kaur S, Singh SP, Elkahloun AG, Wu W, Abu-Asab MS, Roberts DD. tumor metastasis in response to blockade of the chemokine receptor
CD47-dependent immunomodulatory and angiogenic activities of CXCR6 is overcome by NKT cell activation. J Immunol
extracellular vesicles produced by T cells. Matrix Biol 2014;37:49-59. 2009;183(9):5807-15.
21. Long KB, Beatty GL. Harnessing the antitumor potential of macrophages 33. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R.
for cancer immunotherapy. Oncoimmunology 2013;2(12)e26860. Nanocarriers as an emerging platform for cancer therapy. Nat
22. Kim MS, Haney MJ, Zhao Y, Mahajan V, Deygen I, Klyachko NL, et al. Nanotechnol 2007;2(12):751-60.
Development of exosome-encapsulated paclitaxel to overcome MDR in 34. Davis ME, Zuckerman JE, Choi CH, Seligson D, Tolcher A, Alabi CA,
cancer cells. Nanomedicine 2016;12(3):655-64. et al. Evidence of RNAi in humans from systemically administered
23. Yuan D, Zhao Y, Banks WA, Bullock KM, Haney M, Batrakova E, et al. siRNA via targeted nanoparticles. Nature 2010;464(7291):1067-70.
Macrophage exosomes as natural nanocarriers for protein delivery to 35. Batrakova EV, Li S, Brynskikh AM, Sharma AK, Li Y, Boska M, et al.
inflamed brain. Biomaterials 2017;142:1-12. Effects of pluronic and doxorubicin on drug uptake, cellular metabolism,
24. Vilner BJ, John CS, Bowen WD. Sigma-1 and sigma-2 receptors are apoptosis and tumor inhibition in animal models of MDR cancers. J
expressed in a wide variety of human and rodent tumor cell lines. Cancer Control Release 2010;143(3):290-301.
Res 1995;55(2):408-13. 36. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al.
25. Banerjee R, Tyagi P, Li S, Huang L. Anisamide-targeted stealth Minimal experimental requirements for definition of extracellular
liposomes: a potent carrier for targeting doxorubicin to human prostate vesicles and their functions: a position statement from the International
cancer cells. Cancer 2004;112(4):693-700. Society for Extracellular Vesicles. J Extracell Vesicles 2014;3, https://
26. Kim SK, Foote MB, Huang L. The targeted intracellular delivery of doi.org/10.3402/jev.v3.26913.
cytochrome C protein to tumors using lipid-apolipoprotein nanoparti- 37. Finn OJ. Immuno-oncology: understanding the function and dysfunction
cles. Biomaterials 2012;33(15):3959-66. of the immune system in cancer. Ann Oncol 2012;23(Suppl 8):viii6-9.
27. Yang Y, Hu Y, Wang Y, Li J, Liu F, Huang L. Nanoparticle delivery of 38. Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, De Milito A, et al.
pooled siRNA for effective treatment of non-small cell lung cancer. Mol Microenvironmental pH is a key factor for exosome traffic in tumor cells.
Pharm 2012;9(8):2280-9. J Biol Chem 2009;284(49):34211-22.
28. Kooijmans SA, Fliervoet LA, van der Meel R, Fens MH, Heijnen HF, 39. Morishita M, Takahashi Y, Nishikawa M, Takakura Y. Pharmacokinet-
van Bergen En Henegouwen PM, et al. PEGylated and targeted ics of exosomes — an important factor for elucidating the biological
extracellular vesicles display enhanced cell specificity and circulation roles of exosomes and for the development of exosome-based
time. J Control Release 2016;224:77-85. therapeutics. J Pharm Sci 2017;106(9):2265-9.
29. Sena-Esteves M, Tebbets JC, Steffens S, Crombleholme T, Flake AW. 40. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al.
Optimized large-scale production of high titer lentivirus vector Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic
pseudotypes. J Virol Methods 2004;122(2):131-9. cancer. Nature 2017;546(7659):498-503.

You might also like