High‐Throughput Microfluidic‐Mediated Assembly of Layer‐By‐Layer Nanoparticles (2025)

1 Introduction

Spatiotemporal control of drug delivery has been a major objective of new therapeutic platforms.[1] Nanoparticles (NPs) have been extensively studied as drug carriers to alter the pharmacokinetics and pharmacodynamics of therapeutics, with engineering of NP surface chemistry representing an important strategy to optimize delivery of drugs to disease sites and/or avoid off-target tissue uptake.[2, 3] A versatile approach to engineer surface chemistry is via layer-by-layer (LbL) assembly of polymer coatings on nanoparticles. The LbL technique consists of the deposition of nanoscale polymer films on surfaces through alternating adsorption of molecular layers of complementary polymers, most typically polycations and polyanions.[4-7] The resulting multilayer assemblies, which are several nanometers in thickness, enable controlled drug release as well as modulation of surface properties and the resulting interactions with biological interfaces.[4, 8, 9] Based on the versatility of this approach, we and others have successfully demonstrated the utility of LbL-NPs for targeted drug delivery and controlled drug release in many applications.[5, 10, 11] Assembly of polymer films on NPs can yield increased NP plasma half-life and stability, tumor targeting, and control over the subcellular localization of NPs.[8, 12-18] Moreover, LbL-NPs can be used to effectively deliver small-molecule therapeutics[19-27] and nucleic acids,[28-35] which can be loaded into the polymer film in the NP. The high versatility of this platform approach enables the facile assembly of combination therapies within a single particle to target diverse diseases.

A challenge of LbL-NP synthesis is the serial nature of the layer deposition process. While improvements have been made in LbL-NP assembly methods at the lab scale, these approaches have still required multiple purification steps and/or hard-to-scale methods of mixing polymers and NPs. This includes the use of sonication or vortex mixers[5, 36] coupled with centrifugation or membrane-based purification approaches that suffer from potential interactions of NPs with the membrane.[36] In recent years, there have been considerable advancements in implementing streamlined and scalable LbL assembly via fluidics due to its high degree of control and reproducibility.[37-40] However, the methods used have been highly scale-dependent, such as size exclusion of particles from channels, magnetic diversion of particles, or fine-tuned microparticle retention in fluidized beds.[41-46] This scale requirement has precluded many of these LbL techniques from being extended to NPs.[45] Some newer approaches have been applied to nanomaterials but require heating which alters the polymer structure and potentially damages the NP.[47]

Here we demonstrate a novel approach for multilayer assembly on NPs using microfluidics (MCF) mixing technology. MCF is scalable, continuous, and readily implemented under current good manufacturing practices (cGMP) conditions for clinical-grade NP production.[48-54] As a test case for this new manufacturing method, we synthesized LbL-NPs composed of an interleukin-12 (IL-12)-conjugated liposomal core layered with poly(amino acid) polymers, which has shown promising therapeutic efficacy in preclinical mouse models of metastatic ovarian cancer.[8, 55, 56] We show that, by rational selection of polymer-to-NP ratios for surface charge conversion without the addition of excess polymers, this approach enables LbL films to be constructed without the need for time-consuming purification steps, greatly simplifying LbL-NP preparation.

2 Results and Discussion

2.1 Excess Polymer is Required for LbL Assembly Without Particle Aggregation during Batchwise Polyelectrolyte Adsorption

As a model system with relevance for targeted immunotherapy delivery to cancer cells, we studied anionic liposomes that were surface-conjugated with the potent cytokine IL-12 (IL-12 NPs), and subsequently coated by alternating layers of poly(L-arginine) (PLR) and poly(L-glutamate) (PLE). LbL assembly has been typically performed by alternating sequential incubations of the liposomal core particle with excess PLR and PLE in a low ionic strength buffer. Under these typical self-assembly conditions, each step of polyelectrolyte adsorption must be accompanied by extensive washing/purification to remove excess unbound polymer from the coated particles before the next round of adsorption.

To better understand and optimize LbL assembly, we first sought to define how the polymer:particle weight ratio varied during nanolayer deposition. Unlayered anionic IL-12 NPs were incubated with increasing PLR-to-liposome mass ratios, or“weight equivalents” (wt. eq.). Equivalently, purified PLR-coated cationic IL-12-NPs (PLR/NPs) were incubated with increasing PLE wt. eq relative to theliposome lipid mass. After polymer and NP mixing, we characterized the resulting unpurified polymer-NP assemblies' size and charge via dynamic light scattering (DLS) and electrophoretic mobility, respectively. Starting from very low polymer concentrations, increased polymer-to-NP wt. eq. led to increasing NP charge up to a charge neutralization point (isoelectric point), at which particles rapidly aggregated due to a lack of charge-charge repulsion (Figure 1a,b). Further increases in polymer-to-NP wt. eq. led to reductions in particle size as the overall NP charge inverted, and the zeta potential neared a plateau onset point (POP), where complete charge reversal was achieved. Given the known composition of the liposomes used here, we sought to estimate the charge neutralization and POP wt. eq. for PLR onto the IL-12 liposomes based on charge balance and found values of ≈0.05 and 0.1, respectively, which were similar to our experimental results (Figure1a, see Supplemental Information for derivation). Analogously, we further predicted charge neutralization and POP wt.eq. for PLE of ≈0.2 and ≈0.44 which were consistent with our empirical observations (Figure1b). However, increasing the polymer-to-NP wt. eq. beyond the zeta potential POP was required to form polymer-coated NPs with the lowest overall size and polydispersity index (PDI). Due to this reduced size and increased homogeneity, polymer:NP wt. equivalents beyond the POP are used for LbL-NP synthesis.[32, 40, 57-60] These results suggested that excess polymers present during the layering prevent bridging of NPs by the polymer to avoid aggregate formation (Figure1c). Indeed, it has been previously shown that polymer deposition onto NP surfaces is a kinetically controlled reaction and is expected to be quantitative until the POP.[61]

We next sought to assess whether incubation of NPs with excess polymer beyond the POP increased the amount of polyelectrolyte adsorbed to the NPs. Using conditions identified above that allowed polymer adsorption while maintaining a low polydispersity of the resulting NPs, bare IL-12 NPs or PLR/NPs were added to a solution of fluorescently-tagged PLR (0.3wt. eq.) or PLE (1 wt. eq.) under sonication to allow polymer adsorption.[32, 36] The excess polymer was then removed via tangential flow filtration (TFF). This protocol of LbL deposition under sonication followed by TFF is our current best practice for the production of LbL-NPs.[36] As expected, the production of LbL-NPs via the TFF protocol showed the characteristic inversion of surface charge and minor increases in overall size while maintaining overall low PDI as PLR and then PLE were adsorbed (Figure 2a). However, quantification of the amount of polymer bound to the NPs after purification demonstrated that only ≈0.1wt. eq. of PLR and ≈0.5-0.6wt. eq. of PLE were adsorbed (Figure2b). These results confirmed that incubation of NPs with polymer wt. eq. beyond the zeta potential POP (Figure1b,c) does not increase the amount of polymer incorporated into the LbL films.

Based on these results, we theorized that the assembly of LbL-NPs at the POP could allow us to omit purification steps, as no excess polymers should be present in the solution. To better understand the limitations of performing LbL-NP assembly at the POP polymer-to-NP wt. eq., we compared particles generated with POP wt. eq. of PLR or PLE to the standard layering with excess polymers under bath sonication followed by TFF. While we could generate reasonably sized LbL-NPs with POP wt. eq. of PLR in small-scale test batches (<50µL and <1mgmL−1 lipids), a noticeable increase in NP size was observed when layering with a larger batch size of NPs (≈5mg) was attempted (Figure2c). Omitting sonication during the layering led to a further increase in the resulting NP size compared to the standard approach. Similarly, PLE layering with POP wt. eq. (0.5wt.eq. PLE-to-lipids) on purified PLR-NPs resulted in NP aggregation during layering (Figure2d). Thus, large-scale LbL assembly in static solution even in the presence of sonication could only successfully be performed under conditions of excess polymer, requiring subsequent purification steps.

2.2 Microfluidics-Enabled Mixing Generates Homogeneous LbL-NPs Without Intermediate Purification Steps

As polymer wt. eq. beyond POP only increases the number of free polymer chains in the solution without affecting the LbL coating composition, we theorized that mixing limitations during the standard bath sonication protocol lead to the observed NP aggregate formation. To overcome these limitations and manufacture large-scale LbL-NPs without the need for purification, we explored using microfluidic (MCF) channels for the mixing of polymers at their POP wt. eq. with NPs. We used a commercial MCF cartridge with a bifurcating mixer.[48, 50] This MCF chip platform has been developed for lab- and clinical-scale cGMP manufacturing of NPs, making it an ideal candidate for clinical-scale manufacturing of LbL-NPs.[48] For mixing, solutions of liposomes and polyelectrolytes were each introduced into one of two entry flow ports (Figure 3a); the two solution streams converged at the mixer and the mixed sample was collected from the outlet port.

We first evaluated the effect of flow rate through the channels on the adsorption of PLR to anionic IL-12 liposomes, combining PLR and liposomes at the POP. The target was to achieve NPs similar in size to that of the standard optimized bath sonication protocol. Increasing the flow rate led to reduced PLR-NP Z-avg size and PDI (Figure3b) while maintaining charge conversion (Figure3c), demonstrating improved sample homogeneity without impacting polymer adsorption. Importantly, at flow rates of >3 mLmin−1, PLR-NPs were prepared with the same size and PDI achieved by the standard protocol expected for layered, non-aggregated particles. Higher flow rates than 10mL were unachievable due to flow rate limitations of the MCF chip. Nonetheless, the MCF mixing enabled large-scale polymer deposition at the POP wt. eq.

Based on these promising findings, we devised a two-stage microfluidic mixing process designed to allow two rounds of polyelectrolyte adsorption at POP wt. eq., in order to generate LbL-NPs without purification steps. IL-12-liposomes and PLR were combined in a first-stage MCF mixer followed by a 30min incubation step to ensure polymer adsorption and perform quality tests. The output of the first stage and a PLE solution were used as the input in a second-stage mixer (Figure3d). When we performed POP wt. eq. PLR deposition onto IL-12-NPs at increased scales (>1 mL and >5 mg) using MCF, we could readily achieve the target PLR-NP sizes of the standard bath sonication production protocol across multiple batches (Figure3e). Similarly, depositing PLE at its POP wt. eq. onto MCF-generated PLR-IL-12 NPs could yield PLR/PLE IL-12 NPs at equal or smaller sizes than the standard bath sonication protocol with reproducibility across batches (Figure3f). To validate that all polymers were adsorbed on NPs using the MCF protocol, we used fluorescently tagged PLE polymers to assemble PLE/PLR IL-12 NPs using both the standard TFF-based protocol and the new MCF LbL-NPs. We separated out any free polymer in the LbL-NPs using centrifugal filters and, as expected from the POP of ≈ 0.5wt. eq. for PLE, only ≈50% of PLE was bound to TFF-LbL-NPs prior to purification due to the excess polymer used during assembly with 1wt. eq. of PLE. On the other hand, both purified TFF-LbL-NPs and MCF LbL-NPs had >95% of PLE bound to NPs (Figure3g), confirming the lack of free polymer. Analysis of sample morphology via negative stain transmission electron microscopy (TEM) confirmed that both TFF-based and the MCF-based PLE/PLR-IL-12 NPs had a polymer film on the NP based on the altered staining around the liposome (Figure3h). Further, we did not find signs of significant polyplex formation in either LbL-NP formulation on the TEM micrographs (Figure S1, Supporting Information)

To compare the reproducibility between LbL assembly on IL-12 liposomes via either the standard TFF-based approach or MCF, we compiled the data from independent batches of TFF-LbL (n = 10) and MCF-LbL (n = 8) over the course of over six months. As expected, both methods yielded overall similar NPs with Z-avg 110–150nm, zeta potential of ≈−50mV, and PDI ≤ 0.2 (Figure S2; Table S1, Supporting Information). However, MCF-LbL NPs yielded smaller size and PDI with reduced standard deviations, demonstrating increased sample homogeneity and reduction in amount of partially aggregated LbL-NPs.

2.3 MCF LbL-NPs Maintain Desired Particle Properties In Vitro and Maintain IL-12-LbL-NP Efficacy In Vivo in a Metastatic Ovarian Cancer Mouse Model

PLE/PLR nanolayers assembled on IL-12 liposomes have two functions: First, the PLE layer promotes the targeting of the particles to ovarian cancer cells, and second, prevention of NP endocytosis following binding to the cancer cells, leading to high retention of the LbL-NPs on the cancer cell membrane.[8, 55-57] As the process of polymer adsorption is a kinetically controlled assembly dependent on mixing, concentration, shear stress, and ionic strength, it was critical to ensure that MCF-LbL NPs maintained the desired properties of TFF-LbL-NPs.[62-69]

To determine whether PLE/PLR-IL-12-NPs generated via MCF assembly had the expected cell-targeting properties, we dosed a murine metastatic ovarian cancer cell line, OV2944-HM1 (HM-1) with fluorescent NPs prepared using both the traditional bath sonication with TFF process and MCF assembly. We then quantified NP association with the cells after 4 or 24 h. As expected, MCF LbL-NPs and TFF-based LbL-NPs had significantly increased HM-1 cell association compared to unlayered (UL) NPs (Figure 4a). Moreover, MCF-LbL-NPs had equal or better HM-1 association compared to TFF LbL NPs, confirming that MCF layering maintained and potentially improved the desired high ovarian cancer cell affinity. However, the most critical characteristic of PLE/PLR LbL-NPs is its cell-membrane retention which enables high intratumoral extracellular residence time of the cytokine payload,IL-12.[57] Thus, we evaluated the subcellular localization of NPs 24 h after dosing via confocal microscopy. While UL particles were all endocytosed (Figure4b), both TFF-LbL NPs (Figure4c) and MCF-LbL NPs (Figure4d) presented with NPs on the cancer cell membrane, confirming that PLE/PLR-IL-12-NPs retained their cell surface retention properties when prepared via MCF mixing.

Having validated that LbL-NP assembly via MCF maintained the desired cancer cell association properties, we next wanted to validate that the loaded IL-12 maintained a similar bioactivity to that of TFF-based IL-12 NPs. We dosed HEK-Blue IL-12 reporter cell lines with either free IL-12 or IL-12 bound to PLE/PLR-IL-12-NPs generated via the standard TFF-based protocol or MCF. The IL-12 on MCF-LbL maintained its activity like that of the TFF-LbL (Figure 5a,b). We then sought to evaluate the particle performance in vivo in mice bearing peritoneally disseminated metastatic HM-1 tumors. We used the same dosing paradigm shown previously to extend survival in this mouse model with IL-12-loaded PLE/PLR-LbL-NPs (Figure5c).[57] Both TFF-based and MCF-based LbL-NPs were found to have better control of tumor growth based on whole-mouse tumor bioluminescence readings (Figure5d), significantly extending survival of mice compared to either free IL-12 or UL IL-12 NPs (Figure5e).

2.4 MCF LbL-NPs can be Readily Assembled with Varied Polymer Chemistries and NP Core Sizes for Screening of LbL-NPs Interactions

The LbL platform is a modular approach for tuning NP surface chemistries with a wide range of polymer chemistries showing promising results in preclinical models.[8, 70] To validate that MCF assembly of LbL films was amenable to other polymer chemistries, we performed titration experiments of IL-12 loaded PLR-NPs with varying wt. eq. of hyaluronic acid (HA), poly-L-aspartic acid (PLD), or polyacrylic acid (PAA). Each polymer chemistry showed a distinct POP wt. eq. which followed the expected trend of mass charge density (PAA>PLD>HA) for each of the polymers tested (Figure 6a). Importantly, we could employ these POP wt. eq. for each of these varying polymer chemistries to generate monodisperse LbL-NPs using MCF mixing (Figure6b,c).

In addition to varying chemistries, LbL assembly may be performed on differing NP cores.[18, 32] However, little has been explored on the effect of NP size on LbL-NP properties. Thus, we sought to layer carboxy-modified latex (CML) beads with varying core sizes with PLR and PLE. As expected, smaller CML particles required larger wt. eq. to reach the POP given their higher surface area to mass ratios (Figure6d). However, no discernable trend for the plateau zeta potential values could be determined likely due to the effect of NP size and roughness on the apparent zeta potential.[71, 72] Nonetheless, we were able to readily assemble PLR/PLE coated CML beads of varying sizes (Figure6e). TEM confirmed that particles maintained their homogeneity (Figure S3, Supporting Information). When evaluated for their binding toward HM-1 cells via flow cytometry, all LbL-NPs showed an increase in median fluorescence intensity (MFI) compared to UL NPs (Figure6f). Interestingly, we found that increasing UL NP size increased NP association whereas smaller LbL-NPs showed higher association. We theorized that this effect on small NPs was due to their increased surface area which should allow for higher LbL-NP film interaction with cancer cells. Indeed, when we assessed the correlation between the log-fold change in LbL-NP MFI relative to UL-NP MFI and the available NP surface area, we found that the data showed a clear significant correlation (Figure6g).

The experiments above carried out layering in deionized water, but LbL assembly is also sometimes carried out in the presence of salts to create LbL films with thicker layers.[32, 73]We thus tested whether LbL-NPs could be generated in the presence of salt-containing buffers. Here we employed anionic liposomes devoid of IL-12 with a composition optimized for siRNA loading and delivery from LbL-NPs.[32] We first examined varying PLR:liposome mass ratios to find the POP of PLR required for LbL assembly in 25mM HEPES and 20mM sodium chloride (NaCl), a buffer composition previously found optimal for loading of siRNA into LbL films.[32] ≈ 0.15wt. eq. of PLR was required to reach the POP (Figure S4a,b, Supporting Information). We next compared the size and zeta potential of LbL-NP synthesized from small-scale tests under bath sonication to increased bath sonication and TFF to that of MCF LbL-NPs. Similar to IL-12-LbL-NPs, we found that assembly with MCF enabled more homogenous particle formulations (Figure S4c–e, Supporting Information).

3 Conclusion

Layer-by-layer assembly is a promising technique to modulate the surface properties of NPs for the development of therapeutic drug carriers. Here we demonstrated that microfluidic mixing is highly effective for combining polyelectrolytes and NPs, achieving LbL assembly under conditions amenable to scalable, continuous synthesis for clinical-scale manufacturing. This was facilitated by the finding that MCF mixing enables LbL assembly to occur at a polymer-to-NP weight ratio that is devoid of excess polymer, allowing for the production LbL-NPs without time-consuming purification steps and loss of NP yields. Thus this approach readily increases yield, reduces waste polymers, increases throughput, and requires less expensive equipment compared to the prior optimized LbL protocol for NPs based on TFF (summary of comparison between TFF and MCF assembly of LbL-NPs provided on Table S2, Supporting Information). We validated that these process modifications do not alter the properties of LbL-NPs in vitro and in vivo based on the known properties of IL-12 loaded PLE/PLR-IL-12-NPs. Moreover, we show that this approach can be implemented for various LbL-NP film chemistries and employed in NPs of differing core compositions and sizes. With a library of PLR/PLE CML NPs of varying sizes we show that the increase in LbL-NP association relative to its UL-NP was directly correlated to the total available NP surface area.

Taken together, this work provides a simple method to assemble LbL-NPs at various scales and at increased throughput. Moreover, while the focus of the work was on NPs, the MCF-LbL technique presented here may be applied to larger particles. This approach should facilitate both clinical development of LbL-NPs as well as ease the production of libraries of LbL-NPs which can be implemented to screen for desired traits.

4 Experimental Section

Materials

1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (sodium salt) (POPG), 1,2-distearoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (sodium salt) (DSPG), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-[4-(p-maleimidophenyl)butyramide] (sodium salt) (18:1 MPB-PE), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-dibenzocyclooctyl (DOPE-DBCO), and cholesterol were purchased from Avanti Polar Lipids. Poly-L-arginine (PLR) with a molecular weight (MW) of 9.6kDa, poly-L-glutamic acid (PLE) with a MW of 15kDa, and poly-L-aspartic acid (PLD) with a MW of 14kDa were purchased from Alamanda Polymers. Borondipyrromethene tetramethylrhodamine (BDP TMR) azide (Lumiprobe) and BDP 630/650 azide (Lumiprobe) were conjugated to DOPE-DBCO in chloroform to generate DOPE-TMR and DOPE-630/650. Successful conjugation was validated via thin-layer chromatography which indicated <1% free dye. Hyaluronic acid (HA, 20kDa) was purchased from Lifecore Biomedical. Poly-L-acrylic acid (PAA, 15kDa) was purchased from Sigma Aldrich. Yellow fluorescent carboxy-modified latex (CML) beads (Fluorospheres) with diameters of 20nm, 40nm, 100nm and 200nm were purchased from ThermoFisher Scientific.

Recombinant Single-Chain IL-12 Production

Single-chain IL-12 sequence[74] was synthesized as a genomic block (Integrated DNA Technologies) and cloned into a gWIZ expression vector (Genlantis). Plasmids were transiently transfected into Expi293 cells (ThermoFisher Scientific). After 5 days, cell culture supernatants were collected and protein was purified in an ÄKTA pure chromatography system using HiTrap HP Niquel sepharose affinity column, followed by size exclusion using Superdex 200 Increase 10/300 GL column (GE Healthcare Life Sciences). Endotoxin levels in purified protein was measured using Endosafe Nexgen-PTS system (Charles River) and assured to be <5 EUmg−1 protein.

IL-12 Conjugated Liposome Synthesis

Lipid solutions composed of 65 mol% DSPC, 24 mol% Cholesterol, 6 mol% POPG and 5 mol% of MPB-PE were made in chloroform and dried into a thin film using a rotovap, and then further dried overnight in a desiccator. Lipid films were then hydrated at 0.5-1mgmL−1 using pH 5 deionized water and sonicated for 5 min at 65°C then extruded (Avestin Liposofast-50) at 65°C one time through a 100nm membrane then three times through 50nm membranes. Extruded liposomes were added to an ice bath and the pH was adjusted to pH 7.0 with 10mM HEPES buffer prior to the addition of IL-12 containing a terminal cysteine residue at 0.17wt.eq. with liposomes at 0.33mgmL−1. After overnight incubation with IL-12 at 4°C, any remaining maleimides were quenched with 100-fold molar excess of L-cysteine (Sigma) for 1.5h on ice. For fluorescence labeling of liposomes, 0.2 mol% of DSPC content was replaced by either DOPE-TMR or DOPE-630/650. IL-12 concentration was measured via enzyme-linked immunoassay (ELISA) (Peprotech) and lipid content was quantified via fluorescence. IL-12 free liposomes were made with a 33 mol% DSPC, 33 mol% DSPG, and 33 nol% cholesterol and hydrated and extruded as described previously.[32]

Small-Scale Layer-By-Layer (LbL) Assembly

For small-scale (≈50µL) polymer adsorption onto NP surfaces, polymer solution was added to a 1.7mL tube. The container was then placed under bath sonication (Branson 2510) and the NPs were quickly (<1 s) added to the polymer solution. Samples were allowed to incubate for 5 min for analysis. Any subsequent layers were deposited without any purification unless otherwise indicated. For all polymer-to-NP wt. eq. values, only the NP core mass is considered.

Large-Scale LbL Assembly and Purification via Tangential Flow Filtration (TFF)

Assembly of polyelectrolyte layers at larger scale (>1 mL) was performed as described previously.[36] Briefly, unlayered liposomes were added to a solution with 0.3–0.4wt.eq. of PLR relative to lipid in a glass Erlenmeyer flask under sonication. After 30 min incubation on ice, excess PLR polymer was removed by tangential-flow filtration (TFF) through a 100kDa mPES membrane (Repligen) pre-treated with a 10mgmL−1 solution of free PLR. For the terminal PLE layer, purified particles coated with PLR were added to a solution with PLE in a glass flask under sonication at 1wt.eq. of polymer to lipid. Particles coated with both PLR and PLE were then purified by TFF on a separate 100kDa mPES membrane (Repligen) to remove any excess PLE. For layering, a glass Erlenmeyer flask with <30% of its rated volume was used to facilitate homogeneous NP distribution over the polymer solution and minimize the chance of inhomogeneities in the sample

Microfluidics LbL Assembly

Microfluidic mixing was performed on a NanoAssblr Ignite (Precision Nanosystems) mixing chip (NxGenTM cartrides). Luer lock syringes were separately filled with either polymer solution or liposomes and attached to the microfluidics chip. A syringe pump (Pump 11 Elite, Harvard Apparatus) was then used to control the fluid flow rate into the mixing chip. With the exception of the flow rate studies, assembly was performed with flow rates of 9 mLmin−1 (per channel).

Characterization of Particle Preparations

Dynamic light scattering (DLS) and zeta potential measurements were made on a Zetasizer Nano ZSP (Malvern). Nanoparticle micrographs were acquired using Transmission Electron Microscopy (TEM) on a JEOL 2100F microscope operated at 200kV with a magnification range of 10 000–60 000X.

Fluorescent Polymer Synthesis

PLR was reacted with 2 molar equivalents of BDP-Texas Red (TR)-NHS-ester (Lumiprobe) in DMSO catalyzed with one molar equivalent of triethylamine (TEA). The resulting PLR-TR conjugate was purified via Reverse-Phase High Pressure Liquid Chromatography (RP-HPLC) on a Jupiter C4 column (5 µm particles, 300 Å – Phenomenex) using a water:acetonitrile gradient which started at 20% actetonitrile for 5 min, then increased to 35% in a linear gradient until 10 min. Isocratic elution at 35% was performed for 30 min then the elution buffer was increased to 95% to clean out the column for 10 min then dropped back to 20% acetonitrile to re-equilibrate the column for 5 min. After HPLC purification, the polymer-dye conjugated was then lyophilized and stored at -20°C. PLE was reacted with 5 molar excess of sulfo-cy3-NHS-ester (Lumiprobe) in pH 9 PBS (adjusted by adding 0.1M sodium bicarbonate) for 3 h at 25°C then left 18 hr at 4°C. Excess dye was removed via extensive dialysis (3 kDa, Spectrum) against 0.9% NaCl, and then dialyzed against deionized water to remove salts. The resulting PLE-cy3 polymer was lyophilized and stored at -20°C.

Polymer Retention Quantification

To assess the amount of polymer retained on LbL-NPs after TFF processing, IL-12 NPs with 0.2 mol% of DOPE-630/650 was layered with 0.3wt. eq. of a PLR solution composed of 50% PLR and 50% PLR-TR. After purification of excess PLR/PLR-TR via TFF, particles were layered with 1wt. eq. of a PLE solution composed of 50% of PLE and 50% PLE-cy3. After purification of excess PLE/PLE-cy3 via TFF, the sample was diluted 10x into dimethyl sulfoxide (DMSO) to disrupt the NPs. The fluorescence of lipids and polymers were separately quantified on a plate reader (TECAN) compared to standard curves to determine the polymer-to-NP wt. eq. of the purified NPs.

Excess Polymer Quantification

IL-12 NPs were generated via either the standard TFF-based LbL protocol or the MCF protocol, but with the PLE layering solution having 50% PLE-cy3. Free polymers were separated from NPs on a 300kDa centrifugal filter (Vivaspin500, Sartorius) at 30 xg for 20min. Polymer fluorescence in the permeate fluid was then compared to that of the initial sample to determine the fraction of polymer bound to NPs. Permeate DOPE-630/650 fluorescence was measured to confirm complete particle separation.

Assembly of LbL-NPs with Various Outer Layer Polymers

PLR-coated IL-12-NPs were first titrated with increasing concentrations of either HA, PLD, or PAA to determine the POP wt. eq. Then the solution of NP at 1–2mgmL−1 was mixed the polymer at the POP wt. eq. as described in Microfluidics LbL assembly.

Assembly of LbL Films on CML Beads

CML beads of varying sizes were titrated with varying concentrations of PLR to determine the POP wt. eq. Particles were then mixed with the determined PLR wt. eq. as described in Microfluidics LbL assembly. The same process was repeated with PLE to generated LbL-CML NPs.

Cell Culture

OV2944-HM-1 cells were acquired through Riken BRC and were cultured in α-MEM while MC38 cells (ATCC) were cultured in DMEM. Cell media was also supplemented with 10% FBS and penicillin/streptomycin with cells incubated in a 5% CO2 humidified atmosphere at 37°C. All cell lines were murine pathogen tested and confirmed mycoplasma negative by Lonza MycoAlert Mycoplasma Detection Kit.

In Vitro Cellular Association

The day before dosing, HM-1 cells were plated on a tissue-culture 96-well plate at a density of 50k cells per well. The next day, wells were dosed with NPs to 0.05mgmL−1 and left for the target incubation time (4 hrs or 24hrs). For analysis of association, the supernatant was removed from the well and diluted 10X with DMSO. Cells were then washed three times with PBS then dissolved with DMSO. Fluorescence of NPs associated with cells was then normalized to supernatant fluorescence. The relative fluorescence of each formulation was then compared to an unlayered liposome control containing the same fluorophore as described previously.

For analysis via flow cytometry of CML beads, NPs were dosed at the indicated concentrations and allowed to incubate with cells at 37°C for 4 h. Cells were washed with PBS then detached from the plates using 0.25% trypsin and stained with DAPI (15 min incubation) for viability assessment and fixed with 2% paraformaldehyde (30 min incubation) until analysis by flow cytometry using an LSR Fortessa (BD Biosciences).

For confocal imaging, 8-well chambered coverglass (Nunc Lab-Tek II, Thermo Scientific) were treated with rat tail collagen type I (Sigma-Aldrich) per manufacturer's instructions. HM-1 cells were plated onto wells at a density of 10k/well and left to adhere overnight prior to NP treatment. After the desired incubation time with NPs, cells were washed 3x with PBS. After washing, cells were fixed in 4% paraformaldehyde for 10 min then washed (3x with PBS) and stained with wheat germ agglutinin (WGA) conjugated to Alexa Fluor488 (Invitrogen) and Hoechst 33342 (Thermo Scientific) following manufacturer instructions. Images were analyzed using ImageJ. Slides were imaged on an Olympus FV1200 Laser Scanning Confocal Microscope.

Mice

B6C3F1 mice were purchased from Jackson Laboratories. Female mice were used between 8–12 weeks of age unless otherwise noted with weights of 20–25g. All animal work was conducted under the approval of the Massachusetts Institute of Technology Division of Comparative Medicine in accordance with federal, state, and local guidelines.Before mouse treatments or imaging, mice were anesthetized with 2–3% isoflurane.

Efficacy Studies with Metastatic Ovarian Cancer Model

B6C3F1 mice were inoculated intraperitoneally with 106 cells of luc-HM-1-luc in PBS. Five days after inoculation, bioluminescence was measured on an In Vivo Imaging System Spectrum CT (IVIS, Perkin Elmer) 10 min after i.p. injection of 3µg of D-luciferin sodium salt (GoldBio) to confirm tumor engraftment and mice were randomized into treatment groups. At days 7 and 14 post-inoculation, mice were treated intraperitoneally with vehicle (5% dextrose) or 20µg of IL-12 either as a free cytokine, or conjugated to unlayered (UL) NPs, or LbL-NPs. Mice weights were tracked daily after treatments for signs of toxicity. Bioluminescence was tracked for 30 days after tumor inoculation or as needed to evaluate tumor burden.

Statistical Analysis

GraphPad PRISM 10 was used to perform statistical analyses. Comparisons between two groups was performed via unpaired t-tests. For multiple groups or multiple variable analysis, one-way, or two-way ANOVAs were used with Tukey's posthoc correction.

Acknowledgements

I.S.P. and E.G. contributed equally to this work. The authors thank the Koch Institute Swanson Biotechnology Center for technical support. This work was supported in part by the National Institutes of Health (award R01CA235375 to PTH and DJI, and F99CA274651 to ISP), the Marble Center for Nanomedicine, and the Deshpande Center for Technological Innovation. DJI is an investigator of the Howard Hughes Medical Institute. This work was also supported by the Koch Institute Support (core) Grant P30-CA14051 from the National Cancer Institute.

Open Access funding enabled and organized by MIT Hybrid 2025.

    Conflict of Interest

    ISP, PTH, and DJI are inventors on a provisional patent filed by the Massachusetts Institute of Technology related to this work.

    Open Research

    The data that support the findings of this study are available from the corresponding author upon reasonable request.

    References

    High‐Throughput Microfluidic‐Mediated Assembly of Layer‐By‐Layer Nanoparticles (2025)
    Top Articles
    Latest Posts
    Recommended Articles
    Article information

    Author: Trent Wehner

    Last Updated:

    Views: 6846

    Rating: 4.6 / 5 (56 voted)

    Reviews: 95% of readers found this page helpful

    Author information

    Name: Trent Wehner

    Birthday: 1993-03-14

    Address: 872 Kevin Squares, New Codyville, AK 01785-0416

    Phone: +18698800304764

    Job: Senior Farming Developer

    Hobby: Paintball, Calligraphy, Hunting, Flying disc, Lapidary, Rafting, Inline skating

    Introduction: My name is Trent Wehner, I am a talented, brainy, zealous, light, funny, gleaming, attractive person who loves writing and wants to share my knowledge and understanding with you.