Open Access Article
Andrea
Longatti
a,
Christina
Schindler
a,
Andie
Collinson
a,
Lesley
Jenkinson
a,
Carl
Matthews
a,
Laura
Fitzpatrick
a,
Margaret
Blundy
b,
Ralph
Minter
a,
Tristan
Vaughan
a,
Michael
Shaw
cd and
Natalie
Tigue
*a
aDepartment of Antibody Discovery and Protein Engineering, MedImmune Ltd., Granta Park, Cambridge, CB21 6GH, UK. E-mail: tiguen@medimmune.com
bCore Tissue Culture, MedImmune Ltd., Granta Park, Cambridge, CB21 6GH, UK
cNational Physical Laboratory, Teddington, TW11 0LW, UK
dDepartment of Computer Science, University College London, London, WC1 6BT, UK
First published on 16th July 2018
Exosomes are extracellular vesicles that mediate cell-to-cell communication by transferring biological cargo, such as DNA, RNA and proteins. Through genetic engineering of exosome-producing cells or manipulation of purified exosomes, it is possible to load exosomes with therapeutic molecules and target them to specific cells via the display of targeting moieties on their surface. This provides an opportunity to exploit a naturally-occurring biological process for therapeutic purposes. In this study, we explored the potential of single chain variable fragments (scFv) as targeting domains to achieve delivery of exosomes to cells expressing a cognate antigen. We generated exosomes targeting the Her2 receptor and, by varying the affinity of the scFvs and the Her2 expression level on recipient cells, we determined that both a high-affinity anti-Her2-scFv (KD ≤ 1 nM) and cells expressing a high level (≥106 copies per cell) of Her2 were optimally required to enable selective uptake. We also demonstrate that targeting exosomes to cells via a specific cell surface receptor can alter their intracellular trafficking route, providing opportunities to influence the efficiency of delivery and fate of intracellular cargo. These experiments provide solid data to support the wider application of exosomes displaying antibody fragments as vehicles for the targeted delivery of therapeutic molecules.
ulti
esicular
odies (MVBs). During this process cytosolic material is incorporated into the lumen of exosomes, which are subsequently expelled into the extracellular space upon fusion of MVBs with the plasma membrane.2 Once released, exosomes can be taken up by cells in an autocrine, paracrine or endocrine manner.3,4 Several routes by which exosomes are taken up by target cells have been described and include direct membrane fusion or endocytosis via clathrin-mediated endocytosis, macropinocytosis, and caveolae-dependent uptake.5–8 Except for membrane fusion, where the exosome content is released into the cytoplasm directly, whole exosomes enter the recipient cell, whereby cargo release is mediated by fusion of the exosome membrane with the endosome membrane.5,9,10
The capacity of exosomes to transfer biologically active molecules such as RNA, DNA and protein has been extensively studied1,11,12 and strategies to exploit this phenomenon to develop exosomes loaded with therapeutically relevant molecules are now emerging. For example, exosomes can be loaded with protein or RNA cargo by genetic modification of the producer cells, which leads to its incorporation during MVB biogenesis,12 or post-purification by electroporation, detergent permeabilisation, hypotonic dialysis, sonication, transfection or extrusion.13–19 Such “ex vivo” loading strategies have been primarily adopted to encapsulate small molecules, with the observation that the encapsulated cargo demonstrates superior efficacy compared to the free drug, both in vivo and in vitro.19–21
To further exploit the potential of exosomes as therapeutic nanovesicles, a means by which to target exosomes to enable delivery to the desired recipient cell will be important. Targeting mechanisms have been explored previously by displaying naturally occurring peptides or receptor ligands on the surface of exosomes,22 however, such approaches have limitations for universal application. Antibodies or antibody fragments, such as single chain variable fragment (scFv) domains, could provide a versatile alternative for the targeting of exosomes to specific cells because they can be raised to virtually any antigen and they are exquisitely specific. The affinity of an scFv can also be tuned to the desired level, which may be crucial in balancing targeting and exosomal cargo release kinetics.
Most studies to date have incorporated targeting moieties onto the surface of exosomes via genetic modification of cells to encode a fusion to a transmembrane domain that localises to exosomal membranes e.g. Lamp2b.23 In this study we employed a similar strategy; we modified HEK293 cells to express scFv domains with varying affinities for the cell surface receptor ERBB2 (Her2), fused to the C1C2 domain of lactadherin, which peripherally associates with the outer exosome membrane by interaction with phosphatidylserine24 and is highly enriched in exosome fractions.25 We demonstrate that anti-Her2 scFvs can be displayed on the surface of exosomes and confirm that these exosomes can bind to recombinant Her2. In uptake studies, we observed that incorporation of the C1C2 domain at the surface of the exosomes negatively impacts their ability to be taken up by cells, whereas exosomes displaying scFvs with a high affinity for Her2 results in selective uptake into cells expressing a high level of Her2. Furthermore, we show that, by targeting exosomes to specific cell surface antigens via the C1C2 domain, it is possible to alter the routes by which the exosomes are trafficked. Our data suggest that antibody-mediated targeting of exosomes has the potential to be broadly applicable for the development of targeted exosome therapeutics.
When HEK293 cells were incubated with increasing concentrations of CFSE-exosomes we observed that association and uptake of the labelled exosomes into cells, as assessed by flow cytometry, is robust at low concentrations (0.5 × 1011 particles per ml) and is dose-dependent, with no saturation reached even at 3 × 1011 particles per ml during a 4 h incubation (Fig. 1A and ESI Fig. S2A†). Subsequently a time course was performed with 3 × 1011 particles per ml, and exosome association and uptake reached steady-state between 4 and 18 h continuous incubation, before a reduced signal was observed (Fig. 1B), likely due to degradation of the CFSE dye in lysosomes. For all subsequent experiments an incubation period of 4 h was used to achieve robust exosome uptake.
![]() | ||
| Fig. 1 Exosomes derived from HEK293 cells are taken up efficiently by HEK293 cells in a time and dose dependent active manner. (A) HEK293 cells were incubated with increasing amounts of purified CFSE-exosomes, or with PBS, in duplicate for 4 h and recipient cell fluorescence was assessed by flow cytometry. Average mean fluorescence intensity (MFI) values were then calculated and normalised to the MFI value achieved at 3 × 1011 particles per ml. Error bars represent the standard error of the mean (SEM) for three independent experiments. Histograms can be found in ESI Fig. S2A.† (B) HEK293 cells were incubated for increasing amounts of time with 3 × 1011 particles per ml CFSE-exosomes (triangles), or with PBS (open circles) in duplicate and assessed by flow cytometry. MFI values were then calculated and normalised to the MFI obtained at 4 h incubation. Error bars represent the SEM for three independent experiments except for the 15, 30 and 60 minutes timepoints, which were only analysed once. (C) HEK293 cells were incubated with 3 × 1011 particles per ml CFSE-labelled exosomes (green) for 4 h and stained with Alexa Fluor-555 Wheat Germ Agglutinin (WGA; plasma membrane; red) and Hoechst (nucleus; blue) post-fixation, and visualised using confocal microscopy. (D) HEK293 cells were incubated with 3 × 1011 particles per ml CFSE-exosomes (green) for 4 h, then fixed (but not permeabilised) and stained with anti-CD81 to outline the plasma, followed by anti-mouse Alexa Fluor-647, and visualised using structured illumination super-resolution microscopy (SIM). The maximum intensity projection of each axis is shown. (E) HEK293 cells were incubated with CFSE-labelled exosomes or PBS, treated in the presence or absence of proteinase K, on ice or at 37 °C for 4 h, in duplicate. Uptake was assessed by flow cytometry, and the MFI values were normalised to the 37 °C values (second column). Statistical analyses were performed using one-way ANOVA with multiple comparisons, comparing to exosome uptake at 37 °C in the absence of proteinase K (column 2). Error bars represent the SEM of three 3 independent experiments. (F) Fifteen immortalised adherent cell lines were tested for their ability to take up HEK293-derived exosomes. Cells were incubated with 3 × 1011 particles per ml CFSE-labelled exosomes in duplicate for 4h and subsequently labelled with CellMask Deep Red to normalise for cell volume. CFSE-derived MFI values were then calculated and normalised to the CellMask Deep Red signal for each cell line. The values were subsequently normalised to the HEK recipient cell value and plotted. Error bars represent the SEM for three independent experiments. | ||
The intracellular localisation of CFSE-exosomes was confirmed using confocal microscopy where a CFSE-derived signal could be observed in the intracellular space following staining of the plasma membrane with labelled wheat germ agglutinin (Fig. 1C). The intracellular location of CFSE-exosomes was also visualised by super-resolution structured illumination microscopy (SIM) images, which showed an abundance of CFSE-labelled vesicles in the cell lumen (Fig. 1D and ESI Movies S1A and B†).
To confirm that, in the flow cytometry-based assay, we observe active exosome uptake as has been described previously5–7,26 HEK293 cells were incubated with CFSE-exosomes at 37 °C, or on ice for 4 h, and the fluorescence of the recipient cells was monitored by flow cytometry (Fig. 1E). As a control, PBS that had been incubated with an appropriate volume of FBS and CFSE and purified in the same manner as the exosome preps, was used. When the cells were placed on ice, where endocytosis and intracellular transport is inhibited, CFSE-exosome uptake was reduced to 25% of that observed at 37 °C, compared to 10% for the CFSE-only control, suggesting that the majority of CFSE-exosome transfer observed at 37 °C represents active cellular uptake. Consequently, and for simplicity, we have used the term “uptake” when describing subsequent flow cytometry experiments. Furthermore, when exosomes were pre-treated with proteinase K, which removes proteins exposed on the surface of exosomes, a 62% reduction in uptake was observed, indicating that protein(s) on the exosome membrane play a key role in mediating exosome binding and internalisation.
Next, we compared the exosome uptake efficiency of fifteen immortalised adherent cell lines, to identify a cell line that demonstrates efficient exosome uptake for use in subsequent experiments (Fig. 1F). The panel of cell lines was incubated for 4 h with CFSE-exosomes and analysed by flow cytometry. In parallel, CellTracker Deep Red staining was used to determine total cell volume. By normalising the CFSE-derived signal to the CellTracker Deep Red signal, we eliminated bias arising from differences in recipient cell volume, enabling comparisons to made between cell lines. We observed differences in uptake efficiency between the individual cell lines, with the most efficient cell line, Hep3B, demonstrating a 3-fold higher signal compared to the least efficient cell line, Capan-1. Interestingly, none of the cell lines in the panel demonstrated significantly increased exosome uptake compared to HEK293 cells; thus, HEK293 cells were used for all subsequent exosome uptake experiments.
A transgene was constructed to encode one of the anti-Her2 scFvs or an irrelevant scFv, fused at its C-terminus to a myc tag followed by the C1C2 domain of lactadherin (Fig. 2A), which associates with phosphatidylserine, a lipid that has been shown to be enriched in exosomes.24 This fusion protein is preceded by a signal peptide (SP) to direct the fusion protein into the secretory pathway.30 These scFv-C1C2-encoding sequences were incorporated into JumpIn T-REx HEK293 cells to generate isogenic stable cell lines where the transgene is inserted at a single and predefined genomic locus, thereby reducing any positional effects on expression levels between the different cell lines. We validated the exosome-producing cell lines by performing flow cytometry detecting surface-exposed scFv via the myc tag, and demonstrated that each isogenic cell line exhibits a uniform cell population and surface scFv-expression level (ESI Fig. S2B†). We then isolated exosomes from all four cell lines and demonstrated via immunoblotting that fusion with the C1C2 domain enabled the scFv to partition into an exosome preparation as demonstrated by the presence of the exosomal markers CD63 and Alix, and absence of the endoplasmic reticulum contaminant calnexin (Fig. 2B, ESI Fig. S1B†). This immunoblotting experiment also confirmed that the different scFv-targeted exosomes were produced to an equivalent level and purity, suggesting that expression of the scFv-C1C2 fusion protein did not affect key exosome properties. To calculate the number of scFv molecules present in our exosome samples, we performed an immunoblotting-based quantification of the G98A-scFv exosomes compared to a purified myc-tagged scFv. Based on our calculations we estimate an average of approximately 1100–1400 scFv molecules per exosome to be present in our preparations (ESI Fig. S3†).
To confirm that Her2-targeted exosomes could bind to recombinant Her2 we performed an ELISA assay, and demonstrated that the two higher affinity Her2 scFv displaying exosomes (B1D2 and ML39) could bind to Her2 in a titratable manner, whereas the low affinity (G98A) and irrelevant scFv-exosomes exhibited little or no binding, respectively (Fig. 2C).
The uptake of scFv-exosomes into the cell line panel was tested by incubation of purified CFSE-labelled exosomes with cells for 4 h followed by flow cytometry analysis. When the uptake of different exosome preparations was compared between cell lines (Fig. 3B), there was a significantly higher signal observed for the SK-BR-3 cell line compared the other cell lines, for all the exosomes tested; this increase in uptake reached highly significant levels (2–3-fold increase) when Her2-targted exosomes were employed, reflective of the high Her2 level (4.2 × 106 receptors per cell) present on these cells. The HCC1954 cell line also demonstrated a somewhat increased propensity to take up non-targeted exosomes, however, despite displaying a similar level of Her2 on its surface (3.6 × 106 receptors per cell), uptake of Her-2 targeted exosomes was significantly lower than observed for the SK-BR-3 cell line.
By comparing the uptake observed for each cell line between the exosome preparations (Fig. 3C) we observed a universal increase in uptake for the targeted exosomes vs. the irrelevant control, which could reflect the presence of Her2 on the cell surface on all the cell lines tested. For HCC1954, and more strikingly for SK-BR-3, however, there was a statistically significant increase in signal when the two high affinity (ML39 and B1D2) scFv-bearing exosomes were used. Of note, there was no difference in uptake of ML39 and B1D2 exosomes into both HCC1954 and SK-BR-3 cells.
To eliminate any cell line-specific differences in uptake, we created isogenic HEK293 cell lines with different Her2 surface levels. Recombinant human Her2 was stably introduced into HEK293 cells via lentiviral transduction and three clonal cell lines with either low (HEK-Her2-L), intermediate (HEK-Her2-M), or high (HEK-Her2-H) Her2 surface levels were isolated and the number of receptors per cell for each isogenic cell line were determined (Fig. 4A and ESI Fig. S2C†). The HEK-Her2-L cell line has a similar level of Her2 expression to HEK293 parental (1.4 × 105 receptors per cell), the HEK-Her2-M cell line has an intermediate expression level (7 × 105 receptors per cell) and the HEK-Her2-H cell line has expression levels similar to HCC1954 and SK-BR-3 cells (2.8 × 106 receptors per cell). We therefore concluded that these isogenic cell lines would be well suited to study Her-2 targeted exosome uptake.
We investigated the uptake of both wild-type and recombinant (non-targeted and targeted) exosomes into the isogenic HEK293-Her2 cell lines. By comparing the uptake of different exosome preparations between cell lines, we observed a significant increase in uptake with ML39- and B1D2-bearing exosomes into HEK-Her2-H cells, but not into HEK parental or HEK-Her2-L or HEK-Her2-H, suggesting that a threshold of Her2 receptors per cell is required for efficient uptake (Fig. 4B). In agreement with the data obtained with the tumour cell lines (Fig. 3), there was no significant difference in the signal observed between the ML39 and B1D2 exosomes. When we compared the different cell lines, we could see a clear reduction (∼2-fold) in the signal between wild-type (non-modified) and C1C2-scFv bearing exosomes, suggesting that the expression of the C1C2-scFv fusion protein has an impact on binding of exosomes to HEK cells (Fig. 4C). Indeed, this reduction in uptake for C1C2-bearing exosomes was also observed in the tumour cell line panel (ESI Fig. S2E†), indicating that this phenomenon is not restricted to HEK cells. Interestingly, when ML39 and B1D2 exosomes were incubated with HEK-Her2-H cells, the signal obtained was higher than for the wild-type exosomes, whereas the irrelevant scFv-bearing exosome signal was two-fold lower.
We then assessed the contribution of association and uptake versus uptake alone for ML39 and B1D2 exosomes into the HEK-Her2-H cell line, and observed an increased proportion of CFSE signal associated with the recipient cells at 4 °C vs. 37 °C (albeit not significant, or of low significance, respectively), compared to unmodified or irrelevant scFv-bearing exosomes (ESI Fig. S2D†). This indicates that there is a higher proportion of exosomes bound to the cell surface when Her2-targeted exosomes are used.
Finally, the flow cytometry uptake data were verified by high-throughput confocal imaging of the HEK wt and HEK-Her2 cell lines incubated with non-targeted vs. targeted exosomes (Fig. 4D).
In this study, we have assessed the feasibility of using scFvs as bespoke, specific targeting motifs for exosomes. We fused anti-Her2 scFvs to the C1C2 domain of lactadherin and evaluated the impact of different scFv affinities on the ability of the targeted exosomes to be taken up by a panel of tumour cell lines and recombinant Her2-expressing HEK cell lines. To monitor exosome uptake by recipient cells we first established a robust exosome uptake assay that could quantitate binding and uptake of CFSE-labelled exosomes, and used it to optimise exosome concentration, incubation time, and recipient cell type. We observed that exosome uptake was dose-dependent and efficient at concentrations of 0.5–3 × 1011 particles per ml, and reaches steady-state between 4–18 h of continuous incubation of the exosomes with recipient cells (Fig. 1A and B) similar to previous reports.5–7,26,37 We also showed that the majority of the signal measured was intracellular, representing active uptake and internalisation, rather than association with the cell surface alone (Fig. 1C–E).
A comparison of exosome uptake efficiencies across a panel of 15 cell lines identified that HEK293 cells are most efficient at taking up exosomes that have been derived from HEK293 cells, when compared to the other cell lines tested. Indeed, in a similar study using U-87 MG cell derived exosomes, none of the cell types tested (CHO K1, MEF, HUVEC, HeLa) were significantly more efficient at taking up exosomes than U-87 MG cells themselves.26 Although a comprehensive study is lacking, it is interesting to consider that this could be a common phenomenon that points towards a prominent role of exosomes in autocrine and paracrine signalling. In this analysis, we also found that, although all the cell lines demonstrated exosome uptake, some (e.g. Hep3B) are more efficient than others (e.g. Capan-1) (Fig. 1F). These results are in line with other studies that have observed differences in exosome uptake efficiency albeit using different exosome producing cells and different target cells.26,38–41 The ability of cells to take up non-targeted exosomes could reflect their overall endocytic activity or it could be influenced by the presence of different cell surface receptors on the individual cell lines, or a combination of both.
In this study, we generated Her2-targeted exosomes and analysed exosome uptake by both flow cytometry and microscopy-based analyses. First, we assessed the uptake of four different exosome preparations, all expressing an scFv fused to the C1C2 domain of lactadherin (an irrelevant scFv or scFvs of increasing affinity for Her2), into cells endogenously expressing different surface levels of the Her2 receptor. We observed robust uptake of the two high affinity scFv-bearing exosome preparations (ML39; 1 nM and B1D2; 15 pM) into SK-BR-3 cells, as expected based on the high level of Her2 present on these cells. Interestingly, however, HCC1954 cells did not take up targeted exosomes as well as SK-BR-3 cells (Fig. 3B and C) despite similar levels of surface Her2 expression (Fig. 3A); a finding that could be related to the presence of different cell surface receptors (e.g. ERBB3 (Her3) and their influence on Her2 internalisation in the two cell lines).42,43
To eliminate the contribution of such cell line-specific differences to targeted exosomal uptake we generated a panel of isogenic stable cell lines expressing distinct levels of Her2. We found that only in the HEK-Her2-H cell line, expressing 2.8 × 106 receptors per cell, was efficient uptake of the high affinity (ML39 and B1D2) anti-Her2-scFv exosomes observed (Fig. 4B and C); uptake into the HEK-Her2-M cell line (7.5 × 105 receptors per cell) was not achieved. Indeed, for both endogenous and recombinant Her2-expressing cell lines, Her2 levels at or above 2.8 × 106 receptor per cell (HCC1954, SK-BR-3 and HEK-Her2-H) were required to observe Her2 scFv-mediated uptake, and cell-specific differences were encountered. This requirement for high receptor number may limit the choice of therapeutic targets to those that are highly expressed on target cells, and these may not represent the most selectively expressed targets. On the other hand, this result could represent the relatively slow internalisation rate of Her2,44 and targeting a more rapidly internalising receptor (e.g. CD22) could overcome the requirement for high receptor levels and/or high affinity scFvs. In addition, the presence of the receptor at a certain level in the cell may not be sufficient to enable robust uptake, as endocytosis rates and expression of other cell surface receptors may play a role. Certainly, it will be interesting to understand whether exosome uptake can be improved by utilising different scFv-receptor pairings. It could be envisaged, for example, that by displaying scFvs to more than one target on the surface of an exosome, the efficiency and specificity of uptake could be improved. Indeed, it has been demonstrated for a Her2 biparatopic antibody, that the combination of two binding domains to different epitopes on the same receptor can drive rapid internalisation of an otherwise poorly internalising receptor.45
In the uptake experiments, we included non-modified exosomes and, intriguingly, we observed a significantly (∼2-fold) lower signal with C1C2-scFv exosomes, compared to these “wild-type” exosomes, for all the HEK cell lines tested (Fig. 4C). This decrease, also observed in the tumour cell line panel (ESI Fig. S2E†), could be a consequence of the C1C2-scFv domain on the surface of exosomes restricting the ubiquitous interaction of phosphatidylserine with recipient cells (e.g. via binding to TIM proteins on the cell surface), or the masking of other protein–protein interactions required for efficient exosome uptake. The presence of the C1C2 domain also appeared to influence the trafficking route of exosomes, such that the propensity for the exosome preparations to colocalise with ligands that prefer a particular trafficking route was altered (Fig. 5A); wild-type (non-modified) exosomes seem to colocalise with CtxB to a better extent than to Tfn and Dex, whereas those with the C1C2-irrelevant scFv on their surface are found more often co-localised with Dex, which may represent differences in how the exosomes engage with target cells, or downstream intracellular trafficking molecules. It should be emphasized that the colocalisation data presented here, represent the steady-state distribution of the selected ligands after several rounds of uptake, and recycling or degradation. Further longitudinal studies utilising pulse-chase experiments or real-time imaging would be needed to determine the exact uptake and intracellular trafficking routes of targeted and non-targeted exosomes. Nevertheless, the C1C2-based targeting approach appears to simultaneously inhibit ubiquitous uptake, and facilitate selective uptake via specific cell surface receptors, representing an opportunity to improve the therapeutic window of targeted exosome therapies.
Two approaches for the introduction of targeting ligands into exosomes have been described to date; one is based on the non-specific conjugation of ligands (e.g. nanobodies) to proteins on the surface of purified exosomes13 and the other, more commonly described method, involves the genetic engineering of exosome-producing cells to express fusion proteins that are displayed the exosome surface. The fusion partners that have been employed to generate targeted exosomes have included Lamp2b,13,23 PDGFR46 and a GPI anchor signal peptide,47 but their use has not been without limitations. For example, fusion with Lamp2b can lead to cleavage of peptide ligands48 or prevent fusion protein expression entirely (unpublished observations), and the use of a GPI anchor may not provide sufficient ligand density to enable exosome uptake.47 In this report, we used an alternative exosomal motif, the C1C2 domain of lactadherin, to generate targeted exosomes, similar to an approach described previously.24 The C1C2 domain binds to phosphatidylserine which is present on the surface of exosomes,49 and this binding is promoted by increased membrane curvature,49,50 making C1C2 a promising fusion partner for exosomal display. Indeed, characterisation of C1C2-scFv expressing cell lines and the exosomes produced from them, confirmed that the scFv molecules can be detected on the cell surface and are associated with the exosome fractions. In addition, we calculated an average of 1100–1400 scFv molecules per exosome, which compares favourably with the level of the F5 scFv on HER2 antibody-targeted liposomal doxorubicin (MM-302), where a density of 45 copies per liposome was achieved, and suggests that a significant proportion of the exosome surface is decorated with scFv molecules. In a similar exosome-targeting study whereby a GPI anchor-based method was employed to generate targeted exosomes, a value of 0.15–0.25 molecules per exosome was obtained using transmission electron microscopy.47 The number of exosome studies that have calculated this figure is limited, and the most appropriate method for quantification of surface-displayed molecules within exosome populations, or on individual exosomes has not been established, making such comparisons difficult. Nevertheless, fusion to the C1C2 domain appears to represent a very efficient method for the generation of exosomes with a high density of surface-displayed molecules.
Several ligands have been adopted for use as exosomal targeting moieties including peptides such as the GE11 peptide that binds to EGF-receptor,46 protein fragments such as the interleukin (IL)-3 fragment that binds to IL-3 receptor,51 viral proteins and peptides including gp350 of EBV that binds to CD21 on B cells, and a rabies virus glycoprotein peptide that binds to the acetylcholine receptor.23,52 Whilst these approaches provide valuable tools for targeting specific cells, we believe that antibodies or antibody fragments offer many advantages as targeting moieties. Firstly, antibodies can be selected against virtually any target protein or epitope using well-established technologies such as mouse immunisation or phage display.53,54 Secondly, a panel of antibodies to a chosen target will provide varying different affinities which can be optimised or tailored as required. Finally, antibodies demonstrate an exquisite specificity that can be exploited to generate exosomes that can be taken up by chosen cells of interest via binding to a specific cell surface protein. As antibodies require the production and association of two separate heavy and light chains, a single chain variable fragment (scFv), consisting of the variable heavy (VH) and variable light (VL) chains joined by a flexible linker, is often used in applications where only the binding function of the antibody is required. The potential of scFvs as highly specific and versatile exosome targeting motifs, however, has not been fully explored.
Interestingly, there was little difference between the uptake of B1D2 and ML39 exosomes into Her2 positive cells, suggesting that an affinity of 1 nM is sufficient to saturate the exosome uptake mechanism, and that increasing the affinity of the scFv cannot improve this further. This finding, together with the high scFv density on the exosome surface, could suggest that avidity of multiple binding entities has masked the impact of the higher affinity scFvs. Previous studies that have successfully achieved targeting13,23,46 have utilised peptide ligands with affinities for their targets in the same range (iRGD peptide; 17 nM,55 RVG peptide; 1.2 nM56 and GE11 peptide; 22 nM57), however, as the ligand densities were not calculated, and receptor internalisation rates will be different, it is difficult to compare these studies. It could be envisaged that different affinity and avidity constants could impact cargo trafficking downstream of cell surface receptor binding. It is known, for example, that antibodies binding with very high affinity to their target may not be able to dissociate within the endosome, leading to rapid lysosomal degradation.29,58 Extending this phenomenon, it could be envisaged that an internalised exosome that is tightly associated to its target may compromise the natural “back-fusion” process to release its cargo. This is highly relevant to the delivery of loaded exosomes as it would prevent efficient cytoplasmic delivery of the potential therapeutic cargo.
Although it has previously been demonstrated that exosomes can be taken up by target cells via macropinocytosis and clathrin- and caveolin-mediated endocytosis,5–8 the impact of arming exosomes with a targeting mechanism on their trafficking pathway has not yet been explored. In this study, we found that Her2-targeted exosomes showed equivalent co-localisation with Dextran, Tfn and CtxB at steady-state, whereas the non-targeted exosomes showed a preference for Dextran (Fig. 5A). Since it has been reported that Her2 can be taken up via caveolin-1 dependent endocytosis and, to a lesser extent, clathrin-mediated endocytosis,59–62 this difference likely represents anti-Her2-scFv-exosomes binding to Her2 and concomitant internalisation with the receptor. Thus, by directing exosomes to bind to a specific cell surface receptor, we have shown that it is possible to alter the trafficking and likely intracellular fate of the cargo. Targeting receptors that are known to traffic via specific pathways and with predefined kinetics could provide an opportunity to manipulate the fate of potential therapeutic cargo to ensure optimal delivery.
In this study, we have explored the use of scFvs as exosomal targeting motifs. Using Her2 and anti-Her2-scFvs as a model system we have shown that exosomes can be targeted to Her2-expressing cells by genetically engineering the exosome producer cells to incorporate a high-affinity anti-Her2-scFv onto the exosome surface. Targeting exosomes via a specific cell surface receptor may also provide a means to bias the uptake of exosomes towards a more efficient or “productive” intracellular route. If exosomes are to fulfil their promise as natural delivery vehicles for otherwise difficult to deliver therapeutic molecules many hurdles have yet to be overcome, but targeting therapeutic exosomes to a desired tissue will clearly be of high importance in any disease setting. The data presented here provide compelling evidence that scFvs can serve as broadly-applicable targeting motifs for exosomal delivery.
To generate overexpressing Her2 cells, lentiviral particles were first generated. HEK293FT cells were co-transfected with lentiviral vector constructs and pPACKH1 packaging plasmid mix (System Biosciences) using Lipofectamine 2000 (Thermo Fisher Scientific) according to manufacturer's instructions. 48 h post-transfection, culture supernatants containing lentiviral particles were harvested and used to infect HEK293 at 70–80% confluency in media containing 8 μg ml−1 polybrene (Sigma-Aldrich). Transduced cells were selected by addition of 1 μg ml−1 puromycin (Thermo Fisher Scientific) 48 h after transduction. Cells were maintained in selective media throughout. Clonal cell lines representing distinct levels of HER2 expression were isolated by limiting dilution and FACS sorting following cell surface staining using an anti-Her2 antibody (20 μl/1 × 106 cells; Clone Neu 24.7; BD Biosciences, #340553).
For labelling of exosomes, preparations were adjusted to 1e12 ml−1 and labelled with 5(6)-Carboxyfluorescein diacetate N-succinimidyl ester (CFSE; Sigma-Aldrich) at a final concentration of 5 μM in PBS by incubating at 37 °C for 30 minutes followed by purification through three successive Zeba Spin Desalting Columns (7K MWCO, Thermo Fisher Scientific) to remove free CFSE dye. A “PBS control” sample that contained CFSE, PBS and a volume of FBS equivalent to the protein concentration in the exosome preparation was included, to control for protein-bound CFSE that was captured during the purification steps. To determine the effect of removal of exosomal surface proteins on cellular uptake of CFSE labelled exosomes, CFSE labelled exosomes were treated with Proteinase K (Thermo Fisher Scientific) for 15 min at room temperature. Proteinase K action was quenched by addition of an excess of 20 mg ml−1 purified bovine serum albumin (New England Biolabs).
:
1 DPBS and Odyssey blocking buffer (LI-COR Biosciences)). Membranes were probed overnight at 4 °C with antibodies to myc (1
:
1000; Clone 9E10; Invitrogen #13-2500), Alix (1
:
1500; Abcam, #ab88388), CD63 (1
:
1000; Abcam, #ab59479) and Calnexin (1
:
2000; Abcam, # Ab22595), washed and incubated with donkey anti-rabbit IgG IRDye 680 (1
:
10
000; LI-COR Biosciences, #926-32221) and donkey anti-mouse IgG IRDye 800CW (1
:
10
000; LI-COR Biosciences, #926-32212). Immunoblots were washed as above and visualised on the Odyssey CLx Infrared Imaging System.
000 cells per well. For the assay, cells were washed once with Freestyle medium, then 50 μl per well Freestyle medium was added followed by 50 μl per well CFSE-labelled exosomes at 1 × 1012 particles per ml. This is equivalent to approximately 1 × 106 particles per cell assuming the cells have undergone a single population doubling before the assay. For ligand co-localisation studies 50 μl per well Freestyle medium containing 50 μg ml−1 Tfn-AF647, 200 μg ml−1 Dex-AF647 or 0.5 μg ml−1 CtxB (all Thermo Fisher Scientific) was added followed by 50 μl per well CFSE-labelled exosomes at 1 × 1012 particles per ml. After incubation at 37 °C 5% CO2 cells were washed twice with PBS and processed for flow cytometry analysis. For the ice control, immediately after the CFSE-labelled exosome addition plates were placed on ice for 4 h after which cells were washed twice with PBS and processed for flow cytometry analysis. For flow cytometry, cells were dissociated with accutase and washed 3 times in PBS supplemented with 1% BSA. For antibody staining, cells were incubated with anti-Her2-FITC (20μl/1 × 106 cells; Clone Neu 24.7; BD Biosciences, #340553) or anti-myc-FITC (20μl/1 × 106 cells; Clone 9E10, Sigma-Aldrich, SAB4700448) for 30 minutes at 4 °C in the dark and washed 3 times in PBS supplemented with 1% BSA. 5 μg ml−1 DAPI (Sigma-Aldrich) was added to the last wash to assess cell viability. Flow cytometry was performed immediately using an LSR Fortessa cell analyser (BD Biosciences). Data was analysed using the FlowJo version 10 software (TreeStar). For comparison between samples the geometric mean of the median fluorescence intensity (MFI) was determined. At least 10
000 cells were analysed for each sample. Cell debris was excluded by analysis of forward and side scatter area. Cells were gated on single cell populations using forward/side scatter height vs. width and dead cells were excluded by gating on DAPI-negative cells. When different cell lines were compared by flow cytometry, cells were labelled with CellMask Deep Red (Thermo Fisher Scientific) to normalise cell mass after the 4 h incubation with exosomes. Unless otherwise indicated a final concentration of approximately 5 × 1011 particles per ml was used for uptake experiments. Each independent experiment was performed with duplicate samples.
000 cells per well and treated as described above. Cells were fixed with 4% paraformaldehyde (Thermo Fisher Scientific) in PBS and blocked with PBS containing 1% BSA. To label nuclei Hoechst33342 (Thermo Fisher Scientific) was added at 1 μg ml−1 for 2 min. To label the plasma membrane Alexa Fluor-555 conjugated wheat germ agglutinin (AF555-WGA, Thermo Fisher Scientific) was added in PBS/1%BSA at 5 μg ml−1 for 20 minutes followed by 3x washes with PBS/1%BSA. Fixed and stained cells were stored in PBS at 4 °C until acquisition. 2 images per well were acquired at 20x magnification on an ImageXpress Micro widefield high content microscope (Molecular Devices) or 10 images per well were acquired at 60× magnification on a confocal Opera High Content Screening System (PerkinElmer).
Co-localisation analysis was performed as described above but AlexaFluor-647 labelled ligands were added together with exosomes (50 μl at 1 × 1012 particles per ml with 50 μl fresh freestyle medium as above) for 4h at 37 °C (50 μg ml−1 transferrin-AlexaFluor-647, 200 μg ml−1 Dextran-AlexaFluor-647; or 0.5 μg ml−1 CtxB-AlexaFluor-647) (Thermo Fisher Scientific) after which cells were processed as described above for high-throughput confocal microscopy analysis.
Co-localisation was quantified using the Columbus software. Briefly, using the inbuilt software, nuclei were counted based on Hoechst labelling, cytosol was outlined based on wheat germ agglutinin labelling to define the region of interest, and co-localisation in the cytoplasm of each cell was quantified by calculating the Mander's Overlap Coefficients using an unbiased approach based on whole field and local median background subtraction in each image. Three independent experiments were performed where >500 images were analysed for each treatment, in each experiment.
To visualise the intracellular location of exosomes non-permeabilised cells were labelled for 20 minutes with mouse anti-CD81 (clone JS-81, BD Biosciences, #555675), followed by 20 minutes of anti-mouse Alexa Fluor-647 (Thermo Fisher Scientific; both in PBS with 1% BSA with 3× PBS/BSA washes in between). Orthogonal (xy, xz and yz) maximum intensity projection were created from reconstructed focal series of images (z-stacks) to show the 3D position of the exosomes relative to the plasma membrane. Further 3D visualisations of the plasma membrane and exosomes were generated from reconstructed z-stacks using the ImageJ plugin ClearVolume.68
Footnote |
| † Electronic supplementary information (ESI) available. See DOI: 10.1039/c8nr03970d |
| This journal is © The Royal Society of Chemistry 2018 |