Abstract
Nanoparticle (NP) drug carriers have revolutionized medicine and increased patient quality of life. Clinically approved formulations typically succeed because of reduced off-target toxicity of the cargo. However, increasing carrier accumulation at disease sites through precise targeting remains one of the biggest challenges in the field. Novel multivalent ligand presentations and self-assembled constructs can enhance cell association, but an inability to draw direct comparisons across formulations has hindered progress. Furthermore, how nanoparticle structure influences function often is unclear. In this report, we leverage the well-characterized hyaluronic acid (HA)-CD44 binding pair to investigate how the surface architecture of modified NPs impacts their association with ovarian cancer cells that overexpress CD44. We functionalized anionic liposomes with 5 kDa HA by either covalent conjugation via surface coupling or electrostatic self-assembly using the layer-by-layer (LbL) adsorption method. Comparing these two methods, we observed a consistent enhancement of NP-cell association with the self-assembly LbL technique, particularly with higher molecular weight (≥ 10 kDa) HA. To further optimize association, we increased the surface available HA. We synthesized a bottlebrush glycopolymer composed of a polynorbornene backbone and pendant 5 kDa HA and layered this macromolecule onto NPs. Flow cytometry revealed that the LbL HA bottlebrush NP outperformed the LbL linear display of HA. Cellular visualization by deconvolution optical microscopy corroborated results from all three constructs. Using exogenous HA to block NP-CD44 interactions, we found the LbL HA bottlebrush NP had a 4-fold higher binding avidity than the best performing LbL linear HA NP. We further observed that decreasing the density of HA bottlebrush side chains to 75% had minimal impact on LbL NP stability or cell association, though we did see a reduction in binding avidity with this side-chain modified NP. Our studies indicate that LbL surfaces are highly effective for multivalent display, and the mode in which they present a targeting ligand can be optimized for NP cell targeting.
Keywords: nanoparticle, glycopolymer, hyaluronic acid, layer-by-layer, self-assembly, CD44, ovarian cancer
Graphical Abstract

Introduction
Nanoparticle carriers have revolutionized medicine and increased the quality of life for millions of patients. Advantages of Nanoparticle (NP)-based delivery include their high surface area to volume ratios, ability to encapsulate therapeutic agents, and modifiable surfaces that can be decorated with targeting functionality.1, 2 Major FDA approvals highlight the success of this strategy: liposomal-encapsulated antimicrobials like AmBisome3 and Arikayce,4 PEGylated nanocarriers like Doxil5 and Onivyde,6 and, excitingly, solid lipid nanoparticles used in the COVID-19 vaccines by Pfizer7 and Moderna.8 These recent technologies leverage high cargo loading or reduced off-target toxicity for their increased efficacy, while some further take advantage of non-specific accumulation of the carrier at the site of interest.9 Nanoparticles that improve the localization or accumulation of therapeutics at the disease site through specific affinities to cells or tissues will further enhance efficacy and overall patient survival.10
Optimizing NP delivery could be accelerated by understanding how NP surface functionalization influences tissue and cell specificity.11–14 Appending various ligand types, including antibodies,15 peptides,16 and sugars,17 to nanoparticle surfaces can target NPs to cell receptors of interest, especially those upregulated during disease. Analyses of structure-function relationships that assess NP ligand surface presentation and density have revealed differences in binding and accumulation.18–25 These studies underscore the importance of a multivalent presentation, which can augment binding avidity and internalization.26, 27
Covalent attachment of binding epitopes to NPs can be problematic because of the kinetic barriers encountered during heterogeneous reactions. Indeed, covalent conjugation typically leads to a limited number of epitopes appended onto a NP surface.28 By contrast, noncovalent surface adsorption through self-assembly is a robust method to achieve high surface functionalization. One such non-covalent strategy is the layer-by-layer (LbL) electrostatic assembly technique.29–32 In this approach, NPs are assembled via oppositely charged polyions that are sequentially adsorbed onto a charged colloidal NP surface.33 This process facilitates the formation of uniform particles presenting a charged ligand of interest. The identity of the charged group can have a drastic effect on cell-mediated uptake. For example, Correa et. al.34 showed that ovarian cancer cells preferentially take up carboxylate- over sulfate-presenting surfaces. LbL NPs that leverage this surface display have been effective in multiple applications,35 including as dual-targeting agents,36 responsive theranostics,37 imaging modalities,38 and cancer treatments.39
Direct comparisons between types of surface modifications and arrangements—covalent, noncovalent, and ligand density and presentation—for precise targeting are sparse within the NP literature. The lack of information complicates an understanding of NP effects, thereby hindering efforts to tailor NPs for specified functions. The well-characterized hyaluronic acid (HA)-CD44 binding pair is highly suitable for a head-to-head investigation between ligand functionalization and presentation,40–45 as HA binds avidly to the CD44 cell-surface receptor to mediate cell targeting and internalization.46 HA is an anionic glycosaminoglycan that is a suitable polyanion for generating stable LbL NPs that target cell-surface CD44.43 Such NPs have increased circulation half-lives and enhanced tumor accumulation at ovarian cancers overexpressing CD44.34, 47 We therefore sought to investigate how varying the NP surface presentation of HA would affect its specific association and binding avidity with two ovarian cancer cell lines derived from high-grade serous ovarian cancer, the predominant subtype in the clinic.48
To test the influence of surface presentation on NP targeting, we generated three distinct liposome surface modifications with HA: 1) HA covalently coupled; 2) HA deposited electrostatically; and 3) a synthetic high-density HA bottlebrush polymer deposited electrostatically (Figure 1). We then compared how these different HA-decorated NPs interact with ovarian cancer cell lines. In this way, we leveraged polymer chemistry, materials science, and nanomedicine to optimize NP-cell interactions. These comparative studies provide guidelines for enhancing targeted NP-cell association, and lay the groundwork for understanding how NP surface presentation affects targeted cell interactions.
Figure 1.

Schematic illustration of the three distinct hyaluronic acid (HA) nanoparticle constructs. A) HA is coupled to the surface of liposomes via copper-catalyzed azide-alkyne click chemistry or B) self-assembled via the layer-by-layer technique with either linear HA or a synthetic bottlebrush HA. C) Chemical structures of the reducing end of each HA derivative.
Results and Discussion
HA presentation on liposomal surfaces alters NP-cell association
To generate liposomes with HA coupled onto the particle surface, we installed an alkyne handle onto the reducing end of 5 kDa HA via reductive amination. We confirmed that the functionalization occurred with high efficiency using 1H NMR and FT-IR (see Methods). We fabricated liposomes with azide-terminated phospholipids to install the HA-alkyne via a click reaction involving the copper-catalyzed azide-alkyne cycloaddition.49 We elected to use 5 mol% of an azide-modified lipid in the liposomes to have our total surface modification in line with clinically approved formulations (e.g. Doxil).5 Though we were unable to quantify the number of HA molecules conjugated on the liposome, we assume that the binding sites are fully saturated because the conjugation was performed with excess HA and with extended reaction time.
In parallel, we generated HA LbL NPs where the HA is the outermost layer.33 We successfully layered poly-L-arginine (PLR) onto an anionic liposomal core (Figure S1), followed by 5 kDa HA as the terminal layer in our NP constructs. We used 5 kDa HA as an anionic layer to directly compare NPs that present HA polysaccharides of the same molecular weight. The coupled and layered HA constructs yielded stable NPs with relatively similar bulk properties; Z-average diameters ranging from 122 ± 2.8 to 144 ± 1.1 nm, polydispersity indices ranging from 0.130 ± 0.081 to 0.153 ± 0.034, and negative zeta potentials of −18 ± 0.3 mV to −56 ± 1.7 mV, respectively (Figure 2A). The less negative zeta potential of the layered NP is likely due to the underlying cationic charge from PLR. Since 5 kDa HA only has ~10-12 carboxylate functional groups available to convert the surface charge, high weight equivalents of 5 kDa HA were required to achieve stable NPs (vide infra).
Figure 2.

Cellular association of two distinct HA NP surface presentations. A) Size and zeta potential measurements of the covalently coupled and noncovalent, linear layered 5 kDa HA NPs. Flow cytometry was used to evaluate the association of the layered, self-assembled HA NPs with B) COV362 or OVCAR8 ovarian cancer cells relative to the median fluorescence intensity (MFI) of the initial, bare liposomes. Error bars represent standard deviation for N = 3 technical repeat measurements in A, and N = 5 technical replicates for B. Fold increase of liposome MFI is defined as the MFI of each formulation normalized to the liposome MFI at a given timepoint. Kruskal-Wallis non-parametric tests with a false-discovery rate of 0.05 were used, in which * q < 0.05.
We next performed flow cytometry to evaluate the extent of NP-cell association. The covalently coupled HA NPs associated significantly worse than the layered NPs. The layered NPs had superior associated with two immortalized high-grade serous ovarian cancer cell lines with high relative CD44 expression levels:50 COV362 and OVCAR8 (Figure 2B). These two cell lines were chosen since tumors derived from them are known for their metastatic progression, complicating drug delivery.51 We hypothesized that HA covalently appended to NPs will have limited accessibility, and therefore will be less capable of engaging CD44 via multivalent binding. Indeed, the cell association of the surface-coupled HA NPs was only modestly increased (10- and 8-fold at 24 h, respectively) relative to unmodified liposomes.
In contrast, the 5 kDa layered HA NPs had notable, statistically significant increases in association with both cell lines compared to the unmodified liposome or the coupled HA (99- and 22-fold at 4 h, respectively). This effect was magnified with longer incubation times (105- and 74-fold respectively at 24 h). We attribute this difference to the more accessible HA in the layered configuration. Specifically, typical LbL assemblies have polyion loops and tails that extend away from the colloidal surface.52 Thus, the layered LbL assemblies would have a more thorough surface coating and more accessible HA. This results in displaying HA in a configuration that is more likely to bind multiple CD44 receptors with high avidity. Indeed, the crystal structure of HA engaging with CD44 reveals that binding occurs through multiple hydrogen bonding events in a buried binding pocket, giving credence to the requirement of available loops of HA from the NP surface.53 These data highlight the advantages of electrostatic LbL adsorption as a robust multivalent surface functionalization strategy.
Molecular weight of the terminal HA layer affects ovarian cancer cell association of LbL NPs
Our determination that HA presented on the LbL assembly enhanced NP-cell association prompted an investigation of the effects of terminal layer HA molecular weight. To this end, we assembled a panel of layered NPs using HA of increasing molecular weight (ranging from 5 kDa to 100 kDa). Cationic layered PLR-adsorbed liposomes were coated with these varying molecular weights of anionic HA. To determine the HA quantities needed for stable charge conversion, routinely defined as approximately |30 mV|,54 we titrated increasing amounts of HA onto the PLR-layered liposome (Figure 3). All HA polymers above 10 kDa required 0.5 weight equivalents (wt. eq.) to NP to convert the underlying cationic nanoparticle charge to at least −25 mV with diameters < 210 nm (Figure 3A). However, the lowest molecular weight, 5 kDa HA, required higher wt. eq. for similar stability and charge conversion; only by layering with 2 wt. eq. of 5 kDa HA did the charge reach a value of −21.7 mV (Figure 3A). We hypothesize that, while the |30 mV| zeta potential for these NPs was not reached, the highly hydrated HA chains provide additional steric stability for these nanoparticles, even at a lower net charge. The 100 kDa HA NPs tend to have larger hydrodynamic diameters (Figure 3A), most likely due to HA hydration and subsequent swelling, as measured using dynamic light scattering (DLS).
Figure 3.

Comparison of how terminal layer HA molecular weight impacts LbL NP assembly and NP-ovarian cancer cell association. A) LbL NP zeta potential and z-average diameters as a function of wt. eq. for each given molecular weight. Cell association is normalized by fold-increase over bare liposomes in both B) COV362 and OVCAR8 cell lines. Error bars represent standard deviations for N = 3 technical measurements for A, and N = 5 technical measurements for B. N.D. stands for no data. Kruskal-Wallis non-parametric tests with a false-discovery rate of 0.05 were used, in which * q < 0.05, ** q < 0.01, and *** q < 0.001.
We next measured the NP-cell association of the increasing HA molecular weight panel of LbL NPs. NPs layered with HA molecular weights > 5 kDa HA had significantly higher association with both cell lines than the LbL NPs layered with 5 kDa HA (Figure 3B). This trend was apparent at both time points (4 and 24 h incubation periods). The linear HA used in this study is highly anionic at physiological ionic strengths, prompting discussions about its exact surface morphology.55–57 The 5 kDa HA contour length matches its persistence length suggesting this molecular weight HA may have a stiff rod-like morphology.57 Therefore, when low molecular weight HA is displayed on NP surfaces, the HA oligomers may function as consecutively adsorbed segments, also known as trains, which lay “flatly” on the NP surface. This presentation mode would leave fewer HA repeat units free to interact with cell surface receptors, thereby attenuating the benefits of the multivalent nanoparticle surface. Furthermore, previous studies have shown that HA polymers with fewer than 18 repeat units exhibit monovalent and reversible CD44 binding. In contrast, HA composed of more than 18 repeat units can bind to multiple copies of CD44 and therefore interact with higher avidity.41 The 18 repeat units needed for multivalent binding correspond to a molecular weight of roughly 7 kDa, which matches the increased cellular association we observed for HA polymers of 10 kDa and above. This apparent molecular weight threshold may also explain the plateau18 we observed in cellular uptake of NPs layered with higher molecular weights of HA. Once there are HA loops off the NP surface, the layered architecture likely does not change considerably with longer HA polysaccharides. Thus, as long as the NPs are generated using HA of sufficient length (≥ 10 kDa), their cellular interactions are similar.
Bottlebrush glycopolymer HA constructs effectively engage cell surfaces
Our results concerning increased HA binding with greater molecular weight prompted us to test whether an LbL assembly displaying a bottlebrush arrangement of HA would afford even more effective binding. The bottlebrush architecture offers a pendant display of linear HA molecules, which we hypothesized would increase the availability of binding partners relative to NPs displaying linear HA. To enable direct comparisons, we used the previously synthesized HA-alkyne as the side chain by appending it to a poly(norboronene) polymer backbone bearing azide handles. We leveraged the ring-opening metathesis polymerization to generate poly(norboronene) polymers of controllable size and low dispersity.58 We then performed a post-polymerization functionalization involving displacement of the α-chloro amide groups on the polymer with sodium azide to afford the parent polymer (see Supplemental Information). This azide-functionalized polymer was then exposed to the 5 kDa HA-alkyne under standard azide-alkyne cycloaddition click chemistry conditions45, 59 to furnish the target bottlebrush HA polymer. Using this polymer in an LbL NP assembly, we generated monodisperse particles of equivalent size and zeta potential to those previously tested (Figure S2).
We employed time-lapse deconvolution microscopy to visualize cell-association differences in our family of NPs. We labeled COV362 cells with a CellTracker dye that is known to pass through cell membranes. We then monitored uptake of our NPs, imbued with fluorescently tagged lipid cores, up to 30 min post-addition (Figure 4A). Differences in NP interactions with COV362 cells (Figure 4A) were readily apparent. The covalently coupled HA NPs have minimal cell-surface association, whereas the linear HA-layered and bottlebrush HA-layered NPs show improved engagement. The extent of internalization increased over the 30 min time course for both layered HA NP constructs, but little internalization of the covalently coupled HA NPs was observed. In representative cell images taken after one hour (Figure 4B), the bottlebrush layered NPs colocalized with endo-lysosomes. In contrast, little, if any, colocalization of the linear layered particles was observed. These notable results led us to further explore the bottlebrush polymer-modified NPs design space.
Figure 4.

Micrographs obtained from live cell imaging of COV362 cells treated with the three different HA NP architectures. A) First 30 min following NP addition is shown. NPs are pseudo-colored magenta, and Cell Tracker Blue signal, used to visualize the nucleus, is pseudo-colored gray. B) Representative images at 60 min post-NP addition with LysoTracker, used to visualize endo-lysosomes pseudo-colored cyan. Co-localization between the NP and LysoTracker signal is shown in white. Scale bars = 10 μm.
Bottlebrush HA-glycopolymer outperforms linear HA on LbL NPs, providing a modular platform for chemical modification
Given the effectiveness of the HA bottlebrush-coated NPs in previous studies, we investigated how the polymer composition influenced NP-cell association. We synthesized bottlebrush copolymers substituted with a lower mole fraction of HA by decreasing the molar equivalents of HA-alkyne included in the cycloaddition relative to propargyl-PEG3-alcohol (PEG) (Figure 5A). The click reaction proceeded in high yield for both alkyne and PEG, enabling the generation of three bottlebrush copolymers: 3:1, 1:1, and 1:3 HA:PEG (Figure 5A). Decreasing the density of HA in the copolymer led to a concomitant decrease in anionic charge and water solubility. Consistent with the reduced charge density of bottlebrush copolymers by including higher PEG functionalization, the weight equivalents required for stable, charge-converted particles, as indicated by both nanoparticle sizes < 200 nm and zeta potentials < −11 mV, doubled from 0.25 wt. eq. for the 100% HA bottlebrush and 3:1 HA:PEG to 0.5 wt. eq. for the 1:1 and 1:3 HA:PEG bottlebrush (Figure 5B). Similar shifts in zeta potential arise when plotting data in terms of equivalent carboxylate units (Figure S3). Again, we note that the rule of thumb of |30 mV| zeta potential for stable colloids is not reached, probably due to the steric stabilization of the large, surface absorbed bottlebrush polymer.
Figure 5.

Synthesis, particle assembly, and cellular association of bottlebrush HA-layered NPs. A) Synthetic scheme for bottlebrush HA-substituted polymers with PEG side-chain co-addition, with a schematic illustration of the three different bottlebrush compositions as outer layered NPs. B) Bottlebrush LbL NP zeta potential and z-average diameters as a function of wt. eq. at each given bottlebrush formulation. Arrows indicate the ratio at which the LbL NPs aggregated, as defined by > 100 nm increase in Z-average diameter as compared to the parent nanoparticle. Error bars represent standard deviations for N = 3 technical measurements. C) Cell association in median fluorescent intensity normalized to untreated cells for both COV362 and OVCAR8 cell lines. Error bars represent standard deviations for N = 5 technical replicates. Kruskal-Wallis non-parametric tests with a false-discovery rate of 0.05 were used, in which * q < 0.05 and *** q < 0.001. D) Reduction in OVCAR8 association as a function of exogenous 100 kDa HA added. Error bars represent standard deviations for N = 4 technical measurements.
We next evaluated the stability and physiochemical properties of these NPs. NPs layered with 1:1 and 1:3 HA:PEG bottlebrush polymers were unstable after purification. Tangential flow filtration, which was used to purify all other NPs, resulted in aggregation, most likely from the stripping of polymer at high shear rates. Dialysis in a buffered solution also failed to afford stable NPs from these bottlebrush polymers (data not shown). This instability likely arises from their reduced charge density, as seen by the lower net negative zeta potential of the corresponding NPs. With less anionic NPs, electrostatic repulsion is decreased, causing attractive forces to drive aggregation.60 In contrast, layering and purifying the 100% HA bottlebrush and 3:1 HA:PEG bottlebrush polymers resulted in relatively similar size, zeta potential, and stability as linear layered NPs (Figure S2).
Intriguingly, both the layered bottlebrush NPs had a statistically significant higher degree of cell association than the analogous layered linear NPs—both 5 kDa and 40 kDa HA—after 24 hours of incubation (Figure 5C). Furthermore, layered bottlebrush NPs containing no PEG or 3:1 HA:PEG ratio in the HA bottlebrush afforded statistically similar extents of cell association. We postulate that the pendant display of 5 kDa HA from the bottlebrush architecture may allow these small HA chains to act as tails projecting off the NP surface. Both steric and rotational limitations of the poly(norbornene) backbone may hinder the electrostatic coupling of some HA side chains onto the nanoparticle surface and promote their availability for cell surface receptor binding. Thus, NPs presenting ligands on a bottlebrush scaffold can offer increased accessibility and, therefore, more efficient cell targeting.
We note that for the batch of NPs presented in Figure 5, the PLR layer was adsorbed at a lower weight ratio than the previous NPs tested. Previous figures used a 0.3 weight equivalent of PLR, yielding NPs with a highly charged surface of 64.5 ± 1.63 mV. In subsequent batches, a determined, optimal 0.086 weight equivalent of PLR was used, resulting in a less positive surface charge of 45.9 ± 2.27 mV (Figure S1). The reduction in underlying positive charge may enable more HA loops off the NP surface—even for low molecular weight HA—leading to an enhanced relative cellular association. This model is corroborated by the observed NP cellular association plateau with increasing HA molecular weight, suggesting there is a maximum number of available loops off the NP surface above a given molecular weight. These findings further emphasize that linear 5 kDa HA is near the limit of molecular weight for forming stable particles.
Finally, we probed how NP surface architectures might impart increased binding avidity by carrying out competition studies. We incubated OVCAR8 cells with exogenous, high molecular weight HA (100 kDa) for thirty minutes prior to nanoparticle dosing to bind to, and block, cell surface CD44 receptors. We subsequently added each of our nanoparticle formulations to monitor their ability to competitively bind. After two hours of incubation, we observed a decreased cell association across all HA nanoparticle formulations, but no effect in the unfunctionalized, untargeted liposomes (Figure 5D). In accord with the live-cell imaging and flow cytometry data, the bottlebrush HA-layered NPs required the highest concentrations of exogenous HA to outcompete and block nanoparticle-cell associations. These results suggest that multivalent interactions of the dense HA side chains increase the avidity of nanoparticle-cell binding, promoting higher overall association. The effective IC50 values of exogenous HA against bottlebrush HA nanoparticles was higher than that for either the linear HA or 3:1 HA:PEG bottlebrush nanoparticles (Table 1). The presence of neutral PEG side chains for the 3:1 HA:PEG bottlebrush nanoparticles resulted in a reduced avidity compared to linear HA, but higher overall cell association (Figure 5C). Thus, the optimized NPs were highly effective at both associating and avidly binding CD44-positive ovarian cancer cells.
Table 1:
Calculated IC50 values, and confidence intervals (CI), of exogenous HA for blocking OVCAR8 cellular association of each nanoparticle formulation.
| Nanoparticle Formulation | IC50 (μM HA) | 95% CI | 
|---|---|---|
| Liposome | N/A | N/A | 
| 5 kDa Linear HA | 0.85 | 0.57 – 1.3 | 
| 40 kDa Linear HA | 1.5 | 0.99 – 2.3 | 
| Bottlebrush HA | 6.0 | 4.4 – 8.3 | 
| 3:1 Bottlebrush HA:PEG | 0.67 | 0.48 – 0.94 | 
The modularity afforded by this NP design approach presents many opportunities. Specifically, we used PEG within the bottlebrush, but alkynes bearing different functional groups could readily be incorporated. We envision conjugates presenting orthogonal receptor-ligand pairings including neutral moieties, like many saccharides, which could increase the specificity of cell surface targeting. In addition, the layered bottlebrush polymers could be modified to display small molecule fluorophores, drugs, or other therapeutics for imaging or delivery to a tumor.
Conclusion
This direct comparison of NP presentation strategies of a polymeric ligand, covalently coupled or electrostatically absorbed, illustrates that ligand displays are critical in the avidity of receptor-mediated nanoparticle interactions. Stable nanoparticles were assembled when HA was electrostatically layered or covalently coupled, and the association of exclusively layered NPs with target cells increased. Within the linear layered NP design space, cell association was augmented further when 10 kDa molecular weight HA was employed, as this molecular weight was a threshold for nanoparticle stability. We observed the most effective NP-cell association when a synthetic bottlebrush HA polymer was used. Deconvolution optical microscopy of the three distinct modes of presenting HA—covalent coupling, linear layered HA, and layered bottlebrush-substituted HA polymer—corroborated our flow cytometry results, highlighting the best binding NPs were electrostatically layered bottlebrush HA polymer. Microscopy also revealed that the bottlebrush layered NPs had striking colocalization with endo-lysosomes. The formulation using 100% bottlebrush polymer (i.e., no PEG side chains) had the highest binding avidity. Still, our data indicate that up to 25% of the side chains could be non-targeting PEG groups without sacrificing NP stability or cellular association, although somewhat reducing binding avidity. These findings lay the foundation for the generation of multifunctional NPs. Overall, we have introduced a new class of electrostatically self-assembled, multifunctional side-chain nanoparticle constructs for cell targeting, and our studies provide guidelines for further optimizing NP cell association.
Experimental Section
Materials
All chemicals were purchased from Millipore Sigma (Burlington, MA) unless otherwise specified. All sodium hyaluronate molecular weights used, 5 kDa, 10 kDa, 20 kDa, 40 kDa, and 100 kDa, were purchased from Lifecore Biomedical (Chaska, MN). Propargyl-PEG3-alcohol was purchased from BroadPharm (San Diego, CA). Anhydrous solvents were obtained from a solvent purification system by Pure Process Technology (Nashua, NH). Regenerated cellulose dialysis tubing for polymer purification was purchased from Thermo Fisher Scientific (Waltham, MA). Lipids and plant-based cholesterol were purchased from Avanti Polar Lipids, Inc (Birmingham, AL), and the chloroform used to resuspend them was purchased from Macron Fine Chemicals (Randor, PA). Cholesterol was resuspended in a 50 mg/mL solution of chloroform. 1,2-distearoyl-sn-glycero-3-phosphocholine (18:0 DSPC), 1,2-distearoyl-sn-glycero-3-phospho-(1’-rac-glycerol) (18:0 DPSG), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (18:0 DSPE), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-(6-azidohexanoyl) (ammonium salt) (18: azido PE) were resuspended in a 50 mg/mL chloroform solution, 25 mg/mL chloroform solution, 25 mg/mL solution of 65:35 vol:vol chloroform:methanol solution, or a 5 mg/mL of 65:35 chloroform:methanol solution, respectively. Sulfo-Cyanine5 succinimidyl ester dye was purchased from Lumiprobe (Hunt Valley, MD). Membranes used for liposomal extrusion were purchased from Whatman (Maidstown, England). Tangential flow filtration and dialysis membranes for nanoparticle (NP) purification were purchased from Spectrum Labs (Rancho Dominguez, CA), and tubing used for particle purification was purchased from Saint-Gobain (Worchster, MA). 38.5 kDa Poly-L-arginine was purchased from Alamanda Polymers (Hunstville, AL).
Methods
Synthesis of α-alkyne hyaluronic acid (HA-Alkyne)
HA-alkyne was prepared following a modified literature procedure.45 Sodium hyaluronate 5 kDa (1.0 g, 0.215 mmol) was dissolved in acetate buffer pH = 5.6 at 2% w/w (~ 50 mL) and transferred to a round-bottom flask equipped with a stir bar. Propargylamine (1.03 mL, 16.1 mmol, 75 equiv.) was added to the stirring solution at room temperature, followed by sodium cyanoborohydride (1.01 g, 16.1 mmol, 75 equiv.) and the solution was left to stir for at least 5 days at room temperature. The solution was then concentrated and re-dissolved in a minimal volume of water. The aqueous solution was precipitated twice into methanol at −78 °C and then dried under vacuum to afford a white powder in quantitative yield. Representative 1H NMR in D2O is presented in Figure S4. The alkyne is evidenced by the resonance at δ 3.08 ppm (s, 1H).
Synthesis of bottlebrush HA-substituted polymer
Bottlebrush HA was prepared via standard copper-catalyzed azide-alkyne cycloaddition (CuAAC) click chemistry conditions between azide-functionalized polymer P2 (see Supporting Information) and HA-Alkyne. Copper(I) bromide (7.0 mg, 0.048 mmol) was added to a flame dried, 25 mL round-bottom flask equipped with a stir bar under nitrogen atmosphere. The flask was then purged and backfilled with nitrogen three times. In separate flame-dried scintillation vials, azide-functionalized polymer P2 (5.0 mg, 0.024 mmol azide, 0.0016 mmol polymer) and HA-Alkyne (240 mg, 0.048 mmol) were dissolved in a total of 9.7 mL anhydrous dimethyl sulfoxide (0.0025 M azide). These solutions were then injected into the reaction flask and deoxygenated via three consecutive freeze-pump-thaw cycles. Following the final deoxygenation cycle, N,N,N’,N”,N”-pentamethyldiethylenetriamine (10 μL, 0.048 mmol) was injected into the reaction flask, and the resulting solution was a pale teal. The reaction was then heated at 50 °C for at least 48 h. The solution was then transferred to a 10 kDa MWCO regenerated cellulose dialysis membrane and dialyzed against a 1 L acetate buffer at pH = 4.5 to remove both unreacted polymer and HA-Alkyne. The buffer was exchanged at least 5 times before the solution was filtered into tared falcon tubes, frozen with liquid nitrogen, and lyophilized. The product is a fluffy white powder typically obtained in 15% yield and characterized by integrating all 1H NMR signals relative to the new triazole proton at 8.38 ppm. Representative 1H NMR in D2O is presented in Figure S11.
Synthesis of the PEG-HA bottlebrush co-polymer
Copper(I) bromide (10.4 mg, 0.073 mmol) was added to a flame dried 50 mL round-bottom flask equipped with a stir bar under nitrogen atmosphere. The flask was purged and backfilled with nitrogen three times. Azide-functionalized polymer P2 (7.5 mg, 0.036 mmol azide, 0.0024 mmol polymer), HA-Alkyne, and propargyl-PEG3-alcohol were added to three separate flame-dried scintillation vials. The stoichiometry of the alkyne-functionalized molecules was based on the desired extent of hyaluronic acid functionalization. For the 75% functionalization, 136 mg (0.027 mmol) HA-Alkyne and 1.3 mg (0.009 mmol) propargyl-PEG3-alcohol were dissolved in a total of 14.5 mL anhydrous dimethyl sulfoxide (0.0025 M azide), injected into the reaction flask, and deoxygenated via three consecutive freeze-pump-thaw cycles. Following the final deoxygenation cycle, N,N,N’,N”,N”-pentamethyldiethylenetriamine (15 μL, 0.073 mmol) was injected into the reaction flask to afford a pale teal solution. The mixture was heated at 50 °C for at least 48 h. The solution was then transferred to a 10 kDa MWCO regenerated cellulose dialysis membrane and dialyzed against a 1 L acetate buffer at pH = 4.5 to remove the starting materials. The buffer was exchanged at least 5 times before the solution was filtered into tared falcon tubes, frozen with liquid nitrogen, and lyophilized. The product is a fluffy white powder typically obtained in 15–20% yield. The extent of relative hyaluronic acid to PEG3 incorporation was determined by 1H NMR in D2O by integrating the triazole proton at 8.38 ppm relative to overlapped HA α-hydrogen resonances at δ 4.56 and 4.47 ppm. Some of the HA functionalization densities were slightly less than targeted with the copolymer targeting 75% HA, integrating to only 65% HA, and the copolymer targeting 25% HA integrating to 16% HA. 1H NMR spectra for all synthesized copolymers are presented in Figures S12–14.
Liposome formation and conjugation
Liposomal formulation methods have been described previously34 and procedures can be found in the Supporting Information. For the azide liposomes, an additional 5 mol% azidocaproyl PE was added to replace DSPC, giving a molar ratio of 23.3:33.3:5:5:33.4 DSPC:DSPG:DSPE:azidocaproylPE:cholesterol. Liposomes were extruded using an Avestin LiposoFast LF-50 (Avestin, Canada) at 65 °C, and the resulting unilamellar liposomes were checked for uniformity by measuring the size, polydispersity index (PDI), and zeta potential via laser Doppler electrophoresis with dynamic light scattering (DLS) ZS-90 (Malvern, England).
An adapted protocol from Presolski et. al.61 was used to click the HA-Alkyne onto the surface of the azide liposomes. For NPs used in microscopy and flow cytometry, a sulfo-Cy5 dye was conjugated after the initial NP functionalization with the HA-Alkyne via amine acylation with a succinimidyl ester reaction (see Supplemental Information). Conjugated liposomes were purified using the tangential flow filtration (TFF) method described below. Different NP starting batches were used to generate data, but each figure shown was made with a consistent NP batch.
Tangential flow filtration (TFF) for nanoparticle purification
NPs were purified primarily using TFF as previously described.62 Briefly, porous 100 kDa membranes were washed with milliQ water using at least 2x the surface area of the filter in mLs. TFF was performed using a KrosFlo II (Spectrum Laboratories) system and Teflon-coated tubing. Crude LbL NP solutions were passed through these membranes at either 13 mL/min or 70 mL/min for size 13 and size 16 tubing, respectively. Membranes were washed with milliQ water at least 5x the volume of the original solution both before and after NP purification. For layering poly-L-arginine (PLR), membranes were coated with a solution of 0.1 mg/mL PLR for 5 min before NP purificaction.62 Membranes were stored in 70% ethanol to maintain integrity and sterility.
The same procedure was employed to purify Cy5-conjugated liposomes, except that a 10x volume of PBS was used first to remove the dye, as the salt helps to screen the electrostatic interactions. Another 10x volume of milliQ water was used for buffer exchange, and the wastewater was checked for signs of dye, via fluorescent readings, to confirm its removal.
Layer-by-layer assembly
The layer-by-layer assembly was conducted as previously described63 (see Supplemental Information). Terminal-layer bottlebrush HA LbL NPs were dialyzed in 1000x volume in 1 mM HEPES for 72 hours with 3 buffer changes, as the TFF shear force destabilized these constructs (data not shown). Note that ~0.3 weight equivalents (wt. eq.) of PLR was used for the first layer in NPs used for Figures 2 and 3, and ~0.08 wt. eq. of PLR was used for the first layer in NPs used in Figure 4 and 5. Subsequent wt. eq. of linear and bottlebrush HA polymers varied depending on molecular weight, ranging from 0.25 to 2. The final layering buffer for PLR was 25 mM HEPES and 20 mM NaCl at pH 7.4, whereas the layering buffer for linear and bottlebrush HA was 1 mM HEPES at pH 7.4, as the excess salt interferes with deposition of HA as the second layer of these NP.63
Cell culture and flow cytometry
Both OVCAR8 and COV362 cells were grown in 37 °C and 5% CO2, passaged at ~80–90% confluency, and kept below passage number 25. Cell lines were tested monthly for mycoplasma contamination (Lonza MycoAlert kit, LT07–318). OVCAR8 cells were grown in RPMI1640 (Corning) supplemented with 10% FBS (Gibco) and 1% penicillin/streptomycin (pen/strep), and COV362 cells were grown in DMEM (Corning) supplemented with 10% FBS and 1% pen/strep. OVCAR8 and COV362 cells were kindly donated by the Drapkin and Bhatia Labs, respectively.
For flow cytometry experiments determining NP-cell association, 10,000 cells/well were seeded onto a 96-well plate and allowed to adhere for 24 hours. Cells were washed with PBS, then 90 μL of cell culture medium was added. Next, 10 μL of 7 μg/mL NP solution, normalized by fluorescence, was added, and the mixture was incubated for either 4 or 24 hours. Cells were then washed with PBS and lifted by adding 30 μL of trypsin to each well and incubating at 37 °C for 5 minutes. 150 μL of cell culture media was added to each well to quench the trypsin, and the sample was triturated and kept on ice until analysis by flow cytometry (LSR II). FlowJo was used to analyze the resulting data. Only single cell populations, based on untreated cells using the side and forward scatter plots to determine a singlet gate, were quantified for median Cy5 fluorescence intensity on the APC channel (ex. 640, filters 670/30). Analysis of NP intensity was based on this single color without compensation, and represents a measurement of nanoparticle association with the cells.
For competitive binding experiments with exogenous HA, 15,000 OVCAR8 cells/well were seeded onto a 96-well plate and allowed to adhere for 24 hours. Cells were washed with medium and then 90 μL of fresh medium was added containing 100 kDa HA at concentrations ranging from 0 – 200 μM. The mixture was incubated at 37 °C for 30 minutes. After incubation, 10 μL of 7 μg/mL NP solution was added and incubated at 37 °C for 2 hours. Cells were then washed with PBS and lifted by adding 30 μL of trypsin to each well and incubating at 37 °C for 5 minutes. 150 μL of cell culture medium was added to each well to quench the trypsin, and the sample was triturated and kept on ice until analysis by flow cytometry (LSR II). Single cell populations were quantified for median Cy5 fluorescence intensity on the APC channel, as previously described, and normalized to the average MFI of cells that were not pre-treated with exogenous HA. Concentration of inhibitor versus normalized response curves were formed using a least-squares fit with GraphPad Prism to arrive at IC50 values, and confidence intervals, for exogenous HA of each nanoparticle treatment.
Deconvolution Optical Microscopy
A chambered cover glass (Lab-Tek) was coated with rat tail collagen (Corning, 300 μL of 50 mg/mL in 0.02 N acetic acid). After 5 minutes, the wells were washed with room temperature PBS and allowed to dry in a sterile environment. Wells were stored at 4 °C up to one week prior to seeding COV362 cells in 300 μL of medium at 8,000 cells/well. The cells were allowed to adhere for 24 h (37 °C, 5% CO2). Cells were subsequently washed 3x with warm PBS before adding LysoTracker Green (130 nM final concentration, ThermoFisher) and CellTracker Blue CMAC (13 mM final concentration, ThermoFisher) solution, which was prepared right before use in phenol red-free RPMI 1640 (Gibco). Cells were incubated in the dark at 37 °C, 5% CO2 for 45 min, the dye solution was aspirated, samples were washed 3x with warm PBS, and 300 μL phenol red-free RPMI 1640 was added to each well. Immediately before imaging, cells were treated with 15 μL of a 7 μg/mL NP solution (Cy5 channel). The cells were imaged at the designated time points at 37 °C with the Applied Precision DeltaVision Ultimate Focus Microscope with TIRF Module (Inverted Olympus X71 microscope) equipped with 405, 488, 512, and 568 nm lasers. Images were acquired with either a 60x (with enhanced magnification) or 100x objective. All images were acquired with OMX softWoRx software (Applied Precison/GE). Image LUTs were linearly adjusted to improve contrast using FIJI. Z slices were merged into Z projections covering the height of the cell.
Statistical analysis
All statistical analyses were calculated using GraphPad Prism version 8. See figure captions and supplemental information for the details regarding each calculation.
Supplementary Material
Acknowledgments
The authors would like to thank Brandon Johnston for comments on the manuscript. This work was funded in part by the National Institute of Allergy and Infectious Diseases (AI055258 to L.L.K.) and the Koch Institute Support Grant P30-CA14051. E.D.-Y. acknowledges the National Research Foundation, Prime Minister’s Office, Singapore for support under its Campus for Research Excellence and Technological Enterprise (CREATE) programme. N.B. was supported by a Department of Defense Congressionally Directed Medical Research Programs Peer Reviewed Cancer Research Program Horizon Award (W81XWH-19-1-0257) and the NIH-NCI (K99CA255844). A.J.P. would like to thank the Natural Science and Engineering Research Council of Canada (PGS-D). The authors would also like to thank the MIT Koch Institute Swanson Biotechnology Center, which is supported by the Koch Institute Core Grant P30-CA14051. The table of contents graphic, Figures 1, 3, and 5 were all made in part with BioRender.com.
Footnotes
Supporting Information
Supporting information is available free of charge at . Supplemental methods include: small molecule and bottlebrush polymer synthesis; methods for liposome formulation, conjugation, layer-by-layer assembly, and statistics. Figure S1, size and zeta potential measurements of PLR layered nanoparticles; Figure S2, size and zeta potential measurements of nanoparticles layered with HA bottlebrush polymer; Figure S3, zeta potential measurements of HA-PEG copolymers plotted as a function of carboxylate density; Figures S4–8, representative 1H NMR of small molecules; Figures S9–10, representative 1H NMR of parent polymers P1 and P2; Figures S11–14, representative 1H NMR of HA bottlebrush polymers.
References
- 1.Bangham AD; Standish MM; Watkins JC, Diffusion of univalent ions across the lamellae of swollen phospholipids. J. Mol. Bio 1965, 13 (1), 238–IN27. [DOI] [PubMed] [Google Scholar]
- 2.Allen TM; Cullis PR, Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliver. Rev 2013, 65 (1), 36–48. [DOI] [PubMed] [Google Scholar]
- 3.Meyerhoff A, U.S. Food and Drug Administration Approval of AmBisome (Liposomal Amphotericin B) for Treatment of Visceral Leishmaniasis. Clin. Infect. Dis 1999, 28 (1), 42–48. [DOI] [PubMed] [Google Scholar]
- 4.Drug Approval Package: Arikayce Services, U. S. D. o. H. H., Ed. U.S. Food & Drug Administration: Silver Spring, MD, 2019. [Google Scholar]
- 5.Barenholz Y, Doxil® — The first FDA-approved nano-drug: Lessons learned. J. Control. Release 2012, 160 (2), 117–134. [DOI] [PubMed] [Google Scholar]
- 6.Von Hoff D; Li CP; Wang-Gillam A; Bodoky G; Dean A; Jameson G; Macarulla T; Lee KH; Cunningham D; Blanc JF; Hubner R; Chiu CF; Schwartsmann G; Siveke J; Braiteh F; Moyo V; Belanger B; Dhindsa N; Bayever E; Chen LT, NAPOLI-1: Randomized Phase 3 Study of MM-398 (NAL-IRI), with or Without 5-Fluorouracil and Leucovorin, Versus 5-Fluorouracil and Leucovorin, in Metastatic Pancreatic Cancer Progressed on or Following Gemcitabine-Based Therapy. Ann. Oncol 2014, 25, ii105. [Google Scholar]
- 7.Administration, U. F. a. D. FDA approved first COVID-19 vaccine. https://www.fda.gov/news-events/press-announcements/fda-approves-first-covid-19-vaccine.
- 8.Administration, U. F. a. D., Spikevax and Moderna COVID-19 Vaccine. 2022.
- 9.Shi J; Kantoff PW; Wooster R; Farokhzad OC, Cancer nanomedicine: progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17 (1), 20–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mitchell MJ; Billingsley MM; Haley RM; Wechsler ME; Peppas NA; Langer R, Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov 2021, 20 (2), 101–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yoo J; Park C; Yi G; Lee D; Koo H, Active Targeting Strategies Using Biological Ligands for Nanoparticle Drug Delivery Systems. Cancers 2019, 11 (5), 640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fan Y; Moon JJ, Nanoparticle Drug Delivery Systems Designed to Improve Cancer Vaccines and Immunotherapy. Vaccines 2015, 3 (3), 662–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Blanco E; Shen H; Ferrari M, Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015, 33 (9), 941–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Boehnke N; Hammond PT, Power in Numbers: Harnessing Combinatorial and Integrated Screens to Advance Nanomedicine. JACS Au 2022, 2 (1), 12–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Heath TD; Fraley RT; Papahdjopoulos D, Antibody Targeting of Liposomes: Cell Specificity Obtained by Conjugation of F(ab’) to Vesicle Surface. Science 1980, 210 (4469), 539–541. [DOI] [PubMed] [Google Scholar]
- 16.Josephson L; Tung C-H; Moore A; Weissleder R, High-Efficiency Intracellular Magnetic Labeling with Novel Superparamagnetic-Tat Peptide Conjugates. Bioconjugate Chem. 1999, 10 (2), 186–191. [DOI] [PubMed] [Google Scholar]
- 17.Lundquist JJ; Toone EJ, The Cluster Glycoside Effect. Chem. Rev 2002, 102 (2), 555–578. [DOI] [PubMed] [Google Scholar]
- 18.Alkilany AM; Zhu L; Weller H; Mews A; Parak WJ; Barz M; Feliu N, Ligand density on nanoparticles: A parameter with critical impact on nanomedicine. Adv. Drug Deliver. Rev 2019, 143, 22–36. [DOI] [PubMed] [Google Scholar]
- 19.Tassa C; Duffner JL; Lewis TA; Weissleder R; Schreiber SL; Koehler AN; Shaw SY, Binding Affinity and Kinetic Analysis of Targeted Small Molecule-Modified Nanoparticles. Bioconjugate Chem. 2010, 21 (1), 14–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Song E; Gaudin A; King AR; Seo Y-E; Suh H-W; Deng Y; Cui J; Tietjen GT; Huttner A; Saltzman WM, Surface chemistry governs cellular tropism of nanoparticles in the brain. Nat. Commun 2017, 8 (1), 15322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lv S; Sylvestre M; Prossnitz AN; Yang LF; Pun SH, Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chem. Rev 2021, 121 (18), 11653–11698. [DOI] [PubMed] [Google Scholar]
- 22.Ghaghada KB; Saul J; Natarajan JV; Bellamkonda RV; Annapragada AV, Folate targeting of drug carriers: A mathematical model. J. Control. Release 2005, 104 (1), 113–128. [DOI] [PubMed] [Google Scholar]
- 23.Tjandra KC; Thordarson P, Multivalency in Drug Delivery–When Is It Too Much of a Good Thing? Bioconjugate Chem. 2019, 30 (3), 503–514. [DOI] [PubMed] [Google Scholar]
- 24.Poon Z; Lee JB; Morton SW; Hammond PT, Controlling in Vivo Stability and Biodistribution in Electrostatically Assembled Nanoparticles for Systemic Delivery. Nano Lett. 2011, 11 (5), 2096–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Poon Z; Chen S; Engler AC; Lee H.-i.; Atas E; von Maltzahn G; Bhatia SN; Hammond PT, Ligand-Clustered “Patchy” Nanoparticles for Modulated Cellular Uptake and In Vivo Tumor Targeting. Angew. Chem. Int. Ed. 2010, 49 (40), 7266–7270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lee YC; Lee RT, Carbohydrate-protein interactions: basis of glycobiology. Acc. Chem. Res 1995, 28 (8), 321–327. [Google Scholar]
- 27.Kiessling LL; Grim JC, Glycopolymer probes of signal transduction. Chem. Soc. Rev 2013, 42 (10), 4476–4491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stephanopoulos N; Ortony JH; Stupp SI, Self-assembly for the synthesis of functional biomaterials. Acta Materialia 2013, 61 (3), 912–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gero Decher JBS, Multilayer Thin Films: Sequential Assembly of Nanocomposite Materials. 2nd ed.; Wiley-VCH: 2012; p 1112. [Google Scholar]
- 30.Alkekhia D; Hammond PT; Shukla A, Layer-by-Layer Biomaterials for Drug Delivery. Annu. Rev. Biomed. Eng 2020, 22 (1), 1–24. [DOI] [PubMed] [Google Scholar]
- 31.Iler RK, Multilayers of Colloidal Particles. J. Colloid Interf. Sci 1966, 21 (6), 569–594. [Google Scholar]
- 32.Donath E; Sukhorukov GB; Caruso F; Davis SA; Mohwald H, Novel Hollow Polymer Shells by Colloid-Templated Assembly of Polyelectrolytes. Angew. Chem. Int. Ed 1998, 37 (16), 2201–2205. [DOI] [PubMed] [Google Scholar]
- 33.Decher G, Fuzzy Nanoassemblies: Toward Layered Polymeric Multicomposites. Science 1997, 277 (5330), 1232–1237. [Google Scholar]
- 34.Correa S; Boehnke N; Barberio AE; Deiss-Yehiely E; Shi A; Oberlton B; Smith SG; Zervantonakis I; Dreaden EC; Hammond PT, Tuning Nanoparticle Interactions with Ovarian Cancer through Layer-by-Layer Modification of Surface Chemistry. ACS Nano 2020, 14 (2), 2224–2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Correa S; Dreaden EC; Gu L; Hammond PT, Engineering nanolayered particles for modular drug delivery. J. Control. Release 2016, 240, 364–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Boehnke N; Dolph KJ; Juarez VM; Lanoha JM; Hammond PT, Electrostatic Conjugation of Nanoparticle Surfaces with Functional Peptide Motifs. Bioconjugate Chem. 2020, 31 (9), 2211–2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Boehnke N; Correa S; Hao L; Wang W; Straehla JP; Bhatia SN; Hammond PT, Theranostic Layer-by-Layer Nanoparticles for Simultaneous Tumor Detection and Gene Silencing. Angew. Chem. Int. Ed 2020, 59 (7), 2776–2783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dang X; Gu L; Qi J; Correa S; Zhang G; Belcher AM; Hammond PT, Layer-by-layer assembled fluorescent probes in the second near-infrared window for systemic delivery and detection of ovarian cancer. Proc. Natl. Acad. Sci 2016, 113 (19), 5179–5184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sivakumar S; Bansal V; Cortez C; Chong SF; Zelikin AN; Caruso F, Degradable, surfactant-free, monodisperse polymer-encapsulated emulsions as anticancer drug carriers. Adv. Mater 2009, 21 (18), 1820–1824. [Google Scholar]
- 40.Kim H; Shin M; Han S; Kwon W; Hahn SK, Hyaluronic Acid Derivatives for Translational Medicines. Biomacromolecules 2019, 20 (8), 2889–2903. [DOI] [PubMed] [Google Scholar]
- 41.Lesley J; Hascall VC; Tammi M; Hyman R, Hyaluronan Binding by Cell Surface CD44 *. J. Biol. Chem 2000, 275 (35), 26967–26975. [DOI] [PubMed] [Google Scholar]
- 42.Dubacheva GV; Curk T; Auzély-Velty R; Frenkel D; Richter RP, Designing multivalent probes for tunable superselective targeting. Proc. Natl. Acad. Sci 2015, 112 (18), 5579–5584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dreaden EC; Morton SW; Shopsowitz KE; Choi J-H; Deng ZJ; Cho N-J; Hammond PT, Bimodal Tumor-Targeting form Microenvironment Responsive Hyaluronan Layer-by-Layer (LbL) Nanoparticles. ACS Nano 2014, 8 (8), 8374–8382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Eliaz RE; Szoka FC, Liposome-encapsulated doxorubicin targeted to CD44: a strategy to kill CD44-overexpressing tumor cells. Cancer res. 2001, 61 (6), 2592–2601. [PubMed] [Google Scholar]
- 45.Upadhyay KK; Meins JFL; Misra A; Voisin P; Bouchaud V; Ibarboure E; Schatz C; Lecommandoux S, Biomimetic Doxorubicin Loaded Polymersomes from Hyaluronan-block-Poly(γ-benzyl glutamate) Copolymers. Biomacromolecules 2009, 10 (10), 2802–2808. [DOI] [PubMed] [Google Scholar]
- 46.Lesley J; Hyman R, CD44 structure and function. Front. Biosci 1998, 3, d616–30. [DOI] [PubMed] [Google Scholar]
- 47.Deng ZJ; Morton SW; Ben-Akiva E; Dreaden EC; Shopsowitz KE; Hammond PT, Layer-by-Layer Nanoparticles for Systemic Codelivery of an Anticancer Drug and siRNA for Potential Triple-Negative Breast Cancer Treatment. ACS Nano 2013, 7 (11), 9571–9584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lisio M-A; Fu L; Goyeneche A; Gao Z-H; Telleria C, High-Grade Serous Ovarian Cancer: Basic Sciences, Clinical and Therapeutic Standpoints. Int. J. Mol. Sci 2019, 20 (4), 952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wang Q; Chan TR; Hilgraf R; Fokin VV; Sharpless KB; Finn MG, Bioconjugation by Copper(I)-Catalyzed Azide-Alkyne [3 + 2] Cycloaddition. Journal of the American Chemical Society 2003, 125 (11), 3192–3193. [DOI] [PubMed] [Google Scholar]
- 50.Tsherniak A; Vazquez F; Montgomery PG; Weir BA; Kryukov G; Cowley GS; Gill S; Harrington WF; Pantel S; Krill-Burger JM; Meyers RM; Ali L; Goodale A; Lee Y; Jiang G; Hsiao J; Gerath WFJ; Howell S; Merkel E; Ghandi M; Garraway LA; Root DE; Golub TR; Boehm JS; Hahn WC, Defining a Cancer Dependency Map. Cell 2017, 170 (3), 564–576.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lengyel E, Ovarian Cancer Development and Metastasis. Am. J. Pathol 2010, 177 (3), 1053–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hammond PT, Recent explorations in electrostatic multilayer thin film assembly. Curr. Opin. Colloid Interface Sci 1999, 4 (6), 430–442. [Google Scholar]
- 53.Banerji S; Wright AJ; Noble M; Mahoney DJ; Campbell ID; Day AJ; Jackson DG, Structures of the Cd44–hyaluronan complex provide insight into a fundamental carbohydrate-protein interaction. Nature Structural & Molecular Biology 2007, 14 (3), 234–239. [DOI] [PubMed] [Google Scholar]
- 54.Zeta Potential of Colloids in Water and Waste Water. ASTM: 1985; Vol. 4187-82. [Google Scholar]
- 55.Cowman MK; Spagnoli C; Kudasheva D; Li M; Dyal A; Kanai S; Balazs EA, Extended, Relaxed, and Condensed Conformations of Hyaluronan Observed by Atomic Force Microscopy. Biophys. J 2005, 88 (1), 590–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Weigel PH; Baggenstoss BA, What is special about 200 kDa hyaluronan that activates hyaluronan receptor signaling? Glycobiology 2017, 27 (9), 868–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Berezney JP; Saleh OA, Electrostatic Effects on the Conformation and Elasticity of Hyaluronic Acid, a Moderately Flexible Polyelectrolyte. Macromolecules 2017, 50 (3), 1085–1089. [Google Scholar]
- 58.Choi T-L; Grubbs RH, Controlled Living Ring-Opening-Metathesis Polymerization by a Fast-Initiating Ruthenium Catalyst. Angew. Chem. Int. Ed 2003, 42 (15), 1743–1746. [DOI] [PubMed] [Google Scholar]
- 59.Meng X; Edgar KJ, “Click” reactions in polysaccharide modification. Prog. Polym. Sci 2016, 53, 52–85. [Google Scholar]
- 60.Verwey EJW; Overbeek JTG, Theory of the Stability of Lyophobic Colloids. Elsevier Publishing Company: Netherlands, 1948. [Google Scholar]
- 61.Presolski SI; Hong VP; Finn MG, Copper-Catalyzed Azide-Alkyne Click Chemistry for Bioconjugation. Curr. Protoc. Chem. Biol 2011, 3 (4), 153–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Correa S; Choi KY; Dreaden EC; Renggli K; Shi A; Gu L; Shopsowitz KE; Quadir MA; Ben-Akiva E; Hammond PT, Highly Scalable, Closed-Loop Synthesis of Drug-Loaded, Layer-by-Layer Nanoparticles. Adv. Funct. Mater 2016, 26, 991–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Correa S; Boehnke N; Deiss-Yehiely E; Hammond PT, Solution Conditions Tune and Optimize Loading of Therapeutic Polyelectrolytes into Layer-by-Layer Functionalized Liposomes. ACS Nano 2019, 13 (5), 5623–5634. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
