key: cord-0731202-3sqhtww7 authors: Zhang, Guo; Sun, Jiaming title: Lipid in Chips: A Brief Review of Liposomes Formation by Microfluidics date: 2021-11-03 journal: Int J Nanomedicine DOI: 10.2147/ijn.s331639 sha: ce2b24c353084a84148e666a641cff6644dec8aa doc_id: 731202 cord_uid: 3sqhtww7 Liposomes are ubiquitous tools in biomedical applications, such as drug delivery, membrane science and artificial cell. Micro- and nanofabrication techniques have revolutionized the preparation of liposomes on the microscale. State-of-the-art liposomal formation on microfluidic chips and its associated applications are introduced in this review. We attempt to provide a reference for liposomal researchers by comparing various microfluidic techniques for liposomes formation. Liposomes, or lipid vesicles, are artificially synthesized by one or several phospholipid bilayers entrapping an aqueous core, sizing from nanometers to hundreds of micrometers. First discovered in the 1960s, 1,2 liposomes have become a practical tool in drug delivery, 3-6 membrane science [7] [8] [9] or synthesis of artificial cells [10] [11] [12] due to their similarity to cells and natural vesicles and simplicity to form, operate and modify. The overwhelming success of lipid-based mRNA vaccines has brought a glimpse of hope in addressing the COVID-19 pandemic. 13, 14 Being a safe and efficacious carrier, liposomes can protect loaded materials from external degradation and thus are being rapidly developed as a multifunctional vaccine adjuvantdelivery system with a high capability in inducing desired immune responses for cancer treatment and vaccines. [15] [16] [17] [18] Moreover, breakthroughs in lipid-mediated gene delivery techniques such as targeted genome editing have been reported recently, 19, 20 which have ushered in a promising era for the treatment of genetic diseases. Meanwhile, liposomes have become an indispensable tool in membrane protein studies and synthetic organisms, helping to explore new drugs for cancer and other diseases, as well as elucidating complicated cellular functions. [21] [22] [23] The prevalence of liposomes in research facilitates the search for preparation methods in an economical and feasible way. In addition to reverse evaporation 24 and ethanol injection, 25 commonly used production methods include thin-film hydration, 26, 27 extrusion, 28, 29 electroformation, 30, 31 freeze-drying 32, 33 and double emulsion. 34, 35 Though commonly utilized because of their usability, these macroscale techniques are criticized for their inability to achieve precise control over the size and polydispersity of liposomes and insufficient exploitation of reagents and materials. For example, artificial cellular compartments require precise shape and size control to emulate living cells, yet it is extremely challenging (almost impossible) to achieve such precision using macroscale techniques. It has been acknowledged that a smaller liposome size (≤100 nm) may be suitable for enhancing the activity of an antitumor drug encapsulated in liposomes. 36, 37 The production of "limit size" liposomes with diameters less than 50 nm is only feasible using macroscale techniques such as sonication and homogenization; however, microfluidics easily allows scalable production of SUVs with a size of 20-50 nm. 38 Besides, encapsulated materials within liposomes that dissolve in aqueous solution are sometimes exceedingly expensive (such as mRNA), and bulk methods are inevitably prone to poor reproducibility and material waste. The emergence of microfluidic technology solves this predicament, as liposome formation is narrowed down to a centimeter-level chip, greatly reducing the associated cost and improving liposomes' production efficiency and maneuverability. As an updated version of reviews covering a similar topic published earlier, [39] [40] [41] this review focuses on the current state-of-the-art liposomal preparation within microfluidic chips and related applications are introduced. A comprehensive comparative analysis of various methods classified by design rationales is conducted to highlight their advantages and disadvantages. We intend to provide insightful guidance for each method and its specific application by illustrating the strength and limitations in detail. Lipid vesicles can be classified as unilamellar vesicles (UVs), oligolamellar vesicles (OLVs), multilamellar vesicles (MLVs) and multivesicular vesicles (MVVs) according to their lamellarity. 42 Furthermore, UVs can be divided into small unilamellar vesicles (SUVs) with a particle size of <100 nm, large unilamellar vesicles (LUVs) with a particle size between 100 and 1000 nm, and giant unilamellar vesicles (GUVs) with a particle size > 1 μm according to their size ( Figure 1 ). Generally, dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA) techniques are used to evaluate the size distribution of vesicles. 43, 44 Common methods used to determine the lamellarity of liposomes include nuclear magnetic resonance (NMR), 45 smallangle X-ray scattering (SAXS) 46 and transmission electron microscopy (TEM), 47 to mention a few. As a drug delivery system, MLVs and MVVs are more commonly used to achieve long-term drug delivery effects. 48, 49 To effectively and efficiently function, liposomes need to be prepared at a certain size to allow absorption into cells. Size is a critical parameter in determining liposome drug encapsulation, as well as half-life-in circulation and a smaller liposome diameter, which might be favorable in terms of optimizing in vivo drug release. 50 Thus, SUVs are preferred as drug carriers for vaccines and antitumor drugs, to name a few. 36, 51 Since they are similar in size to cells, GUVs are more commonly used in membrane science and artificial cell synthesis. 52 Differences in size result in differences in properties and applications, but monodispersity, one of the main motivators for utilizing microfluidic chips to prepare liposomes, must be ensured to maintain reproducibility. The stability of vesicles is not only affected by the size and lipid composition but also by the physicochemical environment (such as pH and temperature) across the vesicles. 53, 54 For instance, interbilayer-crosslinked MLVs made by Moon et al retained ~95% of their entrapped proteins when stored in PBS at 4°C for over 30 days. 55 Incorporation of pupylation has also become a popular approach used to enhance stability. 56, 57 Compared to polyethylene glycol (PEG)-modified liposomes that could basically be unchanged after 2 months of storage, conventional liposomes lacking PEG-surfactants showed progressive aggregation and precipitation, suggesting PEGmodification improved the vesicles' stability. 57 The surface electrostatic potential of liposomes has been shown to play a key role in the binding constant and is involved in the uptake of active compounds. 58 Generally, charged liposomes are not prone to aggregation due to electrostatic repulsion. 59 The Zeta potential of liposomes, used to approximate the surface charge, can be determined by laser Doppler velocimetry (LDV). 60 Encapsulation efficiency (EE) is defined as the ratio of the drug encapsulated to the initial amount of drug in the system. It can be affected by many factors, such as drug properties, liposome size, and lamellarity and preparation methods. Effective separation of liposomes from the sample is the most critical step in determining EE. Commonly used separation methods for determining EE include centrifugation, dialysis, chromatography and ultrafiltration, to name a few. Quantification can be conducted after the separation and disruption of liposomes. Yet, NMR and fluorometry can directly determine EE without the separation step avoiding errors caused by the process (Table 1) . Yamamoto et al developed nanoparticle exclusion chromatography for determining EE without pretreatment. Only 5 μL of a liposomal suspension and 3 min of analysis time were required to quantify the amount of unencapsulated drugs. 67 Variance in separation methods may lead to different results. Therefore, it is necessary to choose a suitable method to obtain reliable EE. Notably, high EE does not imply better performance. Bioactive molecules such as proteins may be compromised by various physicochemical factors during the encapsulation, resulting in only a few liposomes that ultimately function normally. 39 This method was first proposed by Jahn et al to prepare nanoscale lipid vesicles in microfluidic chips ( Figure 2 ). 68 The central channel of the lipids dissolved in alcohol was vertically connected to the two side channels with aqueous solutions. As the alcohol was mixed and diluted by the aqueous solutions to a critical concentration, the lipids selfassembled into liposomes spontaneously. Hydrodynamic flow focusing (HFF) can prepare monodisperse liposomes, [69] [70] [71] The lipids composition and concentration are also identified as the primary factors influencing the size of the liposomes and polydispersity index. 72, 73 To further study the self-assembly processes of liposomes, Jahn et al further combined an HFF device with propane jet-freezing. Disclike intermediate structures formed at the interface of lipid−alcohol and buffer, revealing more details about the formation of nonequilibrium liposomes. 74 Balbino et al designed a double hydrodynamic focusing (DHF) device by adding an additional pair of channels based on HFF to produce unilamellar cationic liposomes. 75 DHF doubled the production rate compared to HFF and can also adjust FRR to form liposomes possessing a smaller diameter, while the polydispersity index (PDI) was no different from HFF. 71, 76 To further increase the production rate, Hood et al fabricated a three-dimensional microfluidic hydrodynamic focusing (3D-MHF) device using a concentric capillary array consisting of seven identical glass capillaries collectively fused in a circular pattern. 77 Poly(ether-etherketone) (PEEK) tubings served as the lipid feed line and were threaded through the center of the array ( Figure 3A ). The 3D-MHF device enabled the synthesis of nanoscale liposomes with low PDI at a production rate of four orders of magnitude higher than previous HFF methods due to the entirely radially symmetric diffusion of ethanol-lipid solution. FRR and intra-annular capillary orifice size play important roles in the size control of liposomes prepared by 3D-MHF. However, the usability was compromised due to the complex assembly process of capillary arrays. Soon after, a vertical flow focusing (VFF) device was introduced by the same team. This device was made of a precisely aligned multilayer thermoplastic device with surprisingly low aspect ratios of 1:100 ( Figure 3B ). Monodisperse liposomes (80 to 200 nm in diameter) were prepared at production rates as high as 95 mg/h using this method. 78 Based on the VFF design, Michelon et al and Chen et al used soft lithography or 3D printing to fabricate similar microfluidic chips for high-throughput synthesis of liposomes, respectively. 79, 80 Although using PMDS to fabricate VFF is a clever alternative to enhance the throughput of liposome synthesis, patterning microscale features becomes increasingly difficult as the aspect ratio increases in conventional planar microfabrication processes like soft lithography. Therefore, the throughput of liposomes was curbed as maximum aspect ratios were constrained to 6:1 in the study by Michelon et al. 79 The VFF device International Journal of Nanomedicine 2021:16 manufactured by Hood et al overcomes these limitations and is more impressive and practical. Meanwhile, Chen et al successfully fabricated VFF chips using stereolithography, but the minimal widths of the channel were only at 200 μm due to the limited resolution of the commercial printer. 80 Still, it is believed that 3D printing technologies offer great potential for realizing future improvements in tunable and high-throughput synthesis of nanoscale liposomes. Considerable efforts have been made to utilize HFF design to generate liposomes for drug delivery. For example, liposomes encapsulating plasmid DNA synthesized by HFF could efficiently deliver genes into HeLa cells. 81 Ran et al successfully prepared liposomes of various formulations by HFF, which were then further studied for physicochemical properties. 82 Although high-throughput HFF-based methods like VFF can perform rapid and robust large-scale preparations of liposomes in chips, they suffer from unavoidable low EE. It was reported that EE of both methotrexate-loaded liposomes prepared by DHF or β-carotene-loaded liposomes prepared by VFF were around 60%. 79, 83 Hood et al integrated purification and remote drug loading elements into the HFF device. Though this device reduced the total on-chip residence time of drug-loaded liposomes to 3 minutes unprecedentedly, EE was approximately 72%, which was less compared to conventional protracted remote loading (>95%). 84, 85 Since the formation of vesicles is determined by mixing and diluting solvent into the aqueous solution, solvents such as ethanol cannot be completely removed from liposomes, which might affect the stability of the membranes. Last but not least, HFF-based devices, while practical, show limitations in terms of the sizes and amounts of material generated. For instance, liposomes with diameters less than 50 nm produced by HFF are only achieved at flow rate ratios of 30 or higher, resulting in substantial material dilutions. 70 The emergence of a microfluidic staggered herringbone mixer (SHM) could solve the above problem. Similar to HFF, liposomes can also be formed by using a microfluidic micromixer. As a powerful and general approach to generate liposomes with smaller sizes, SHM designed by Stroock et al was used to mix streams of flows effectively in microchannels. The special staggered herringbone patterns on the channel floor greatly improve mixing efficiency, which can be altered by the asymmetry of the herringbones and the number of herringbones per half cycle. 86 Zhigaltsev et al pioneered SHM ( Figure 4A ) as a scalable manufacturing method to produce limit size lipid They demonstrated the excellent ability of nanoscale liposomes synthesized by SHM to load and retain doxorubicin and confirmed the bilayer structure of SUVs by 31 P NMR and cryogenic TEM (cryo-TEM). 87 They further reported that by using SHM, limit size SUVs with a diameter of around 20 nm could be produced at FRR of only 3 (1.5 mL/min for the flow rates of the aqueous channel). Kastner et al found that the increase of FRR results in increased PDI but decreased size of liposomes prepared by SHM. 88 However, the results of Maeki et al showed that the size and PDI of liposomes prepared by SHM decreased with the increase of FRR simultaneously. 89 Besides, the SHM cycle numbers and the position of the first SHM are also significant factors for the production of vesicles. For example, the size of the liposomes gradually increased with decreasing the SHM cycle numbers and increased distance from where the first SHM was located. Due to insufficient mixing of the aqueous and lipid solutions observed under the high FRR condition, Maeki et al assumed that rather than complete mixing of the solutions, the rapid dilution of ethanol to a critical concentration was essential to form vesicles. 89 In addition to the parameters of SHM itself, the lipid composition, 90 concentration 91 and even solvent selection 92 are all considered key parameters in liposomes formation. It should be pointed out that not all LNPs prepared by SHM constitute a continuous bilayer that qualifies them as liposomes; some LNPs are micelle-like structures (micelles) with a non-aqueous core. 93 Unfortunately, techniques such as cryo-TEM were unaffordable or just unreported in some studies to confirm the structure of LNPs prepared by micromixer. According to those reported results, the formation of liposomes or micelles in SHM might depend mainly on lipid composition and synthesis parameters. 87, 90, 92, 94 Instead of SHM, Kimura et al designed an invasive lipid nanoparticle production (iLiNP) device consisting of 20 sets of zigzag-shaped mixer structures ( Figure 4B ). iLiNP devices enabled the LNPs size tuning at 10 nm intervals in size ranging from 20 to 100 nm precisely by various baffle parameters, flow rate and FRR, which could not be achieved by conventional SHM. 95 Notably, encapsulated materials such as mRNA and siRNA dissolved in aqueous solution are expensive so the LNP must be produced at low and constant FRR conditions. iLiNP device could produce LNPs in size ranging 20-40 nm at flow rates of 50, 100, and 500 μL/min and an FRR of 9. To further enhance the speed and homogeneity of the mixing, Rasouli et al introduced an acoustically driven microfluidic micromixer ( Figure 4C ). 96 The acoustic microstreaming reinforced by integrating sharp edges and bubbles in this device triggered higher throughput of liposomes with lower PDI. Compared to passive micromixers, the intense and pervasive acoustic micro-streams in this device prevented the formation of large nanoparticle aggregates and clogging of the channel. However, the vibrational amplitude of sharp edges rapidly diminished as the flow rate increased, limiting the throughput. Besides, the fabrication of the whole device was somehow complicated for non-professionals; thus, the usability was compromised. Subsequently, the same team applied an electrohydrodynamic micromixer to prepare liposomes with a narrower size distribution at a throughput of 1~2 × 10 10 min −1 ( Figure 4D ). 97 The liposome size was determined mainly by the initial lipid concentration and the FRR in this device. Compared to conventional micromixers that require soft lithography to fabricate grooves, this novel device is easier to implement with only an additional metal deposition step. It is worth mentioning that although the electric field can avoid contaminant clogging or liquid leakage, liposome electroporation, as well as lipid oxidation may occur, affecting the size and stability of liposomes and even the passive permeability of the lipid bilayers. 98, 99 SHM can apparently produce high quantities of limit size liposome/LNPs at lower FRR, avoiding substantial material dilutions and waste. However, problems with relatively low EE have been reported. In the studies by Joshi et al, EE of liposomes prepared by SHM loading a hydrophilic drug (metformin) or a lipophilic drug (glipizide) was around 20% or 40%, respectively. When used to encapsulate highly lipophilic propofol, EE was approximately 40% which is significantly higher than EE in liposomes prepared via sonication (15%). 100 It is worth mentioning that LNPs with micellar structures produced by SHM had higher EE when encapsulating biological molecules such as siRNA. 101 Belliveau et al used a device with six parallel SHM elements to produce limit size LNPs composed of 1-palmitoyl-2-oleoyl-sn-glycero -3-phosphocholine (POPC)/cholesterol to encapsulate siRNA at an extraordinary throughput (72 mL/min or 580 mg LNPs/min), while EE of siRNA was in excess of 95% and PDI was below 0.1. 102 These LNPs exhibited an electron-dense core in cryo-TEM, suggesting micellar structures. Such morphology was distinctly different from bilayer LNPs, which exhibited an electron-dense ring and less dense interior associated with unilamellar vesicle systems with an aqueous interior. It was further proved by Leung et al that high siRNA encapsulation levels for siRNA in LNP systems were inconsistent with bilayer structure since encapsulation was dependent on the presence of cationic lipid. 103 Generally, to prepare liposome/LNPs for drug delivery, micromixers may be a better choice because of their relatively high EE, easy usability and full use of encapsulated materials. The throughput of micromixers had been exponentially scaled up by incorporating an array of numerous mixing channels that operate simultaneously. 104 In fact, commercial devices based on micromixers such as NanoAssemblr™ platforms have been universally used in many studies to prepare liposome/LNPs for different applications, such as CRISPR-Cas9 genome editing for cancer therapy 105 and in utero mRNA delivery for monogenic fetal diseases. 106 Notably, COVID-19 vaccine nanoparticles manufactured by Pfizer are reported to be prepared by microfluidic mixers. 93 Unlike HFF and micromixers, discrete droplets with defined sizes generated by immiscible fluids serve as templates for the formation of liposomes in this method. Generally, these liposomes with diameters measured in micrometers are more used in membrane science and artificial cell research in contrast to drug delivery. Weiss et al developed an approach that merges vesicles and water-in-oil (W/O) droplets to generate cell-like compartments. 107 In this method, LUVs prepared by extrusion were first encapsulated in copolymer-stabilized W/O droplets. By introducing Mg 2+ solution during droplet production, a contiguous lipid bilayer at the inner interface of the droplet was formed due to the transformation of the encapsulated LUVs, which they named droplet-stabilized GUVs (dsGUVs). Assembled lipid compartments could be released from the droplets into the aqueous phase by another microfluidic device ( Figure 5 ). Before demulsification, various proteins and molecules could be loaded sequentially into dsGUVs using pico-injection technology (inspired by Weitz lab designs) 108 to form synthetic cells. By combining different microfluidic technologies, copolymer-stabilized W/O droplets were used as templates for high-throughput preparation of GUVs (10 3 /s) with unique and precise compositions. This elegant technique ensured high EE, and no traces of remaining oil could be detected in the released GUVs under a Raman microscope. A bottom-up reconstitution of the actin cytoskeleton, microtubules or even FoF1-ATP synthase within dsGUVs was successfully obtained by this technology, overcoming fundamental limitations for the design of complex synthetic cells. The same team further reduced the diameter of dsGUVs to ~2 μm by a microfluidic droplet splitter, paving the way towards a targeted delivery of advanced cargos such as microparticles, viruses or macromolecular DNA-robots. 109 They investigated interactions between differently charged GUVs and cells and proved small GUVs to be a practical alternative for transporting and delivering very large, complex cargos such as baculovirus to specific cells, which is not possible by conventional SUV-based delivery. The problem was that droplet-splitted GUVs was not mechanically stable enough, thus leading to a low yield of daughter GUVs. Nonetheless, their sharp control over the GUVs composition and functionalization allowed the construction of GUVs with functional diversity and high targeting specificity. However, the shortcomings of single emulsion templates include the challenging requirement for soft lithography (the minimum width of a microchannel of droplet splitter is less than 2 μm) and the special demand of pico injection devices. First presented by Sugiura et al, 110 lipid-coated ice droplet hydration method was used to prepare monodisperse W/O droplets between 4 and 20 µm in diameter as templates. After freezing, the droplets precipitated and separated, followed by the replacement of hexane solution containing lipid. Next, the solvent was evaporated, while the water droplets were still frozen, and an aqueous solution was added, resulting in the formation of oligolamellar or multilamellar giant vesicles. However, UVs must be yielded by extrusion in this method, and EE decreases from 35% to 12% after extrusion, which restrains the practicability. 110 This method was applied successfully for the entrapment of an enzyme (α-chymotrypsin), but the problem of low DovePress EE was not solved since partial leakage from the initially formed droplets could not be prevented. 111 Pautot et al first described the droplet emulsion transfer method. 34, 112 W/O emulsion prepared by stirring the mixture of aqueous solution and lipid suspension was again transferred to the aqueous phase. Droplets in the emulsion pulled the second monolayer from the oil-water interface to complete the bilayer, resulting in the formation of UVs ( Figure 6A ). They successfully encapsulated various macromolecules like G-action into GUVs as microbioreactors with up to 98% EE. Asymmetric GUVs with a controllable composition of each leaflet independently could also be prepared using this preparation technique. However, these GUVs were polydisperse since the emulsion was prepared by stirring. Further refinements utilizing microfluidic devices then solved the problem ( Figure 6B ). Monodisperse GUVs could be prepared from lipidstabilized W/O droplets using microfluidic droplet formation techniques such as T-junction and flow focusing. However, droplets must be recollected to transfer across the oil-water interface. [113] [114] [115] Importantly, it was reported by Pautot et al that only a fraction of the larger emulsion droplets would pass through the interface to form liposomes because most were likely to break at the interface. 112 They also observed that asymmetric vesicles required several hours of equilibration time to achieve complete lipid coverage of the second monolayer to convert droplets into unilamellar vesicles. Unfortunately, shorter equilibration times (<2 h) led to insufficient coverage and uncontrolled lipid composition, while longer equilibration times (>3 h) resulted in an accumulation of lipid in multilayer structures, creating a crucial limitation in the technique. Successful integration of the generation and transfer of droplets into a microfluidic chip was achieved by Matosevic et al ( Figure 6C ). 116 They used a triangular post to drive lipid-stabilized droplets to transfer through the interface resulting in the deposition of a second lipid monolayer. However, assembly yield was only 5% as droplets were easily lost due to premature contacts with the aqueous flow. EE also decreased to 83% consequently. Karamdad et al further optimized the device to enhance liposome yield and EE. 117, 118 The device contained a micro-step junction manufactured by double-layer photolithography, where W/O droplets were transferred across the oil-aqueous interface to construct monodisperse asymmetric vesicles at high throughput ( Figure 6D ). It is worth noting that double-layer photolithography requires high professional skills. In addition, the Paegel group cleverly integrated a static array on a microfluidic chip for lipid-stabilized W/O droplets trapping. 119 By introducing oils and water alternately, each oil/water boundary crossing would deposit a new monolayer of lipids on the immobilized droplets, suggesting that a phospholipid membrane could be assembled layer-by-layer on these droplets using this platform ( Figure 6E ). Asymmetric multi-bilayers vesicles were successfully assembled within this chip, and this was considered a new tool for studying lipid biosynthesis enzymes and transporters. Based on droplet emulsion transfer, Morita et al developed the droplet-shooting and size-filtration (DSSF) method to prepare monodisperse GUVs (10-20 μm) with controlled asymmetric leaflets in lipid bilayers. 120 Polydisperse W/O droplets were generated from the tip of a glass capillary by centrifugation, and only 10-20% droplets with the smallest diameter were transferred through the oil-water interface selectively by kinetic size filtration (Figure 7 ), as previously observed by Pautot et al. 112 A 1 μL sample solution was sufficient to generate hundreds of liposomes by DSSF without any wasted material. However, a limited amount of solution in the capillary makes it difficult to prepare liposomes efficiently. In addition, residual oil between the leaflets was confirmed, though the authors claimed that it was low enough not to affect the physicochemical and biochemical properties. 120, 121 The main superiority of the droplets emulsion transfer method based on microfluidic devices is the preparation of monodisperse GUVs with asymmetry and high EE, making it suitable for membrane science as well as encapsulation of a diversity of bioactive macromolecules. Adir et al prepared GUVs by the emulsion transfer method and constructed a cell-free expression system by encapsulating bacterial lysate inside GUVs. The obtained synthetic cells were then used to investigate protein/RNA production and activity in an isolated environment. 122 However, the problem of equilibration time, as discussed above, is still unsolved, and controversies arise over the presence of oil residues between leaflets of liposomes since droplets are transferred from oil to the aqueous phase. Kubatta et al demonstrated that an oil layer was accumulated at the top of the giant vesicles (3-5 mm) prepared by bulk transfer method, and oil remnants were also observed in the bilayer by Hu et al 114, 123 Yet the results of the study by Matosevic et al and Pautot et al disagreed since they both detected no trace of oil the bilayer by a thin-layer chromatography analysis or quantitative chemical probing, respectively. Inhibition of membrane protein insertion and function was also not observed. 112, 119 Squalene was recommended by Pautot et al due to a good continuous phase of emulsion and is immiscible in lipid bilayers. 112 Nevertheless, the possibility of oil residues cannot be excluded due to the sensitivity limit of the techniques they used. It seems that different protocols could lead to contradictory conclusions. Imaging based on cryo-TEM or highly sensitive techniques, such as Raman microscopy, is required to verify membrane morphology. First proposed by Shum et al, they prepared lipidstabilized water-in-oil-in-water (W/O/W) double emulsion droplets using glass microcapillary devices as templates. The inner phase consisted of an aqueous solution containing materials used for encapsulation. The middle phase was phospholipids dissolved in a mixture of toluene and chloroform, while the outer phase was poly(vinyl alcohol) (PVA) and glycerol solution ( Figure 8A ). 124 As the solvent layer became thinner during evaporation, the phospholipids were concentrated and then forced to rearrange on the double emulsion templates, thereby forming GUVs. Although monodisperse GUV with defined size and high EE could be prepared by this method, a small amount of organic solvent remained between lipid leaflets after dialysis. To eliminate the presence of undesirable solvents, the same team used another glass capillary microfluidic device to produce W/O/W droplets with ultrathin oil shell (<1 μ m) as templates to prepare GUVs containing minimal residual solvent within their membrane ( Figure 8B ). 125, 126 This time, the inner phase was replaced by PEG and PVA solution, and the oil phase was a mixture of chloroform and hexane. Dewetting occurs since the chloroform evaporates more rapidly than the hexane. The hexane floated as an oil pocket at the top of droplets then evaporated eventually, thus forming GUVs with little residual oil. 126 GUVs prepared by this device were used as a cell-free platform for synthesizing green fluorescent proteins, suggesting that PVA in the inner aqueous phase might not influence the bioactivity of liposomes. 127 Using this technique, a therapeutic enzyme (Cu, Zn-Superoxide dismutase) was successfully encapsulated into liposomes to treat a mice ear edema model. An 7402 of 59 ± 6% and an enzyme activity of 82 ± 3% were obtained. 128 Double emulsion droplets were also generated in PDMS chips. [129] [130] [131] Unlike the capillary microfluidic device, these W/O/W droplets comprised of an internal phase of an aqueous solution, a middle phase of oleic acid and an outer phase containing ethanol ( Figure 8C) . Notably, ethanol served to extract the oleic acid from the middle layer and then evaporated away at last. GUVs are formed by self-assembling of the phospholipids at the oil/ water interfaces. Most GUVs prepared by this method remained stable for a long time (>3 months) and could be used for cell-free protein expression as synthetic cells. 129 There might be residual oleic acid or excess phospholipid left in the lipid shell, but this method was more biocompatible than those described above since toxic solvents such as chloroform were not used. Also, this device did not require any sophisticated design principles and treatments other than an extra step of hydrophilisation of the external channel. The double emulsion templates method can also be used to prepare asymmetric UVs. Arriaga et al further upgraded the glass microcapillary devices. Sophisticated water-in-oil-in-oil-in-water (W/O/O/W) triple emulsion droplets were used as templates for continuous highthroughput preparation of vesicles with asymmetric membranes in a single step ( Figure 8D ). 132 Meanwhile, Lu et al. designed an integrated PDMS chip to fabricate asymmetric UVs. By their ingenious design, lipidstabilized W/O droplets were first formed at the first flowfocusing region, then the oil/inner-leaflet-lipid solution that surrounded the droplets was replaced with the oil/ outer-leaflet-lipid solution. After W/O/W droplets formed in the second flow-focusing region, asymmetric vesicles were collected off-chip followed by the extraction of oil between leaflets of the double emulsions ( Figure 8E ). 133 The oil phase was also extracted by ethanol in this technique, indicating residual oleic acid or excess phospholipid might remain within the lipid shell. It was reported that over 80% of the liposomes remained stable for at least 6 weeks, and the membrane asymmetry was maintained for over 1 day. Compared to W/O/O/W emulsion templates, this method seems easier to handle and produces asymmetric vesicles with monodisperse sizes, high EE and high throughput. However, the choice of solvents was limited to PDMS chips. For example, a solvent typically used in glass capillary devices, chloroform would dramatically swell PDMS and create blockages in the microchannel network. 134 While these results may increase the utility of vesicles with high EE as model biological membranes or for drug delivery applications, there are still some constraints in terms of usability. For instance, the preparation of liposomes by this technique requires careful handling and assembly of multiple glass capillaries or PDMS chips and precise control of several microfluidic parameters simultaneously. Five pumps were used to operate the glass capillary device at the same time in the study by Arriaga et al, not to mention the demanding selections of lipid solvents. 132 Secondly, multiple components like PEG, PVA and Pluronic F-68 are also introduced into the aqueous solution during the formation, which may affect the biological properties of liposomes. Importantly, residual solvent remained after the formation of liposomes. To resolve the oil residues in liposomes once and for all, Deshpande et al developed a microfluidic technique, namely, octanol-assisted liposome assembly (OLA), to form unilamellar, monodisperse, cell-sized GUVs with an efficient, autonomous and fast solvent-extraction mechanism ( Figure 9 ). 135 They replaced the conventional oil phase with 1-octanol. Within a few seconds after its generation, the W/O/W droplet developed into an inner aqueous core entrapped by a lipid bilayer and an outgrowing pocket of 1-octanol containing excess lipids, which completely separated from the liposome in a few minutes. The unilamellarity of liposomes formed by OLA was confirmed by the insertion of the bilayer-spanning protein pore α-hemolysin. The non-leaky detachment of the oil phase also ensured excellent EE. Moreover, the biocompatibility of OLA-based liposomes was verified by encapsulating bacterial divisome proteins. 135, 136 Similarly, Deng et al induced spontaneous and complete dewetting of W/O/W droplets to prepare oil-free monodisperse UVs using a surfactant-assisted microfluidic strategy with coaxial glass capillaries. 21, 137 A triblock copolymer surfactant, Pluronic F-68, was added into the outer phase to control the developing interfacial energies, prompting the oil shells to gradually dewet from the interior drops; therefore, liposomes were separated from oil pockets by excess lipids ultimately just like OLA. Floating oil droplets can be easily removed by pipetting or evaporating after injecting the mixtures into the sucrose solution. Multicompartment liposomes could be formed by varying flow rates with an additional droplet generator ( Figure 10 ). This work is the first to realize the controllable production of multicompartment liposomes in microfluidics. However, the lipid was dissolved in a toxic organic solvent mixture of chloroform and hexane here. Despite concerns that the external aqueous environment where liposomes are collected might be contaminated due to the rupture of oil pockets, and some 1-octanol that might remain in the bilayer, OLA addresses the limitations of other methods to a considerable degree, such as the time-consuming solvent-extraction and remnants of organic solvents in the lipid bilayer. Since the organic solvent used is bio-friendly, OLA is becoming a versatile platform for drug delivery, micro-reactors and synthetic cells. For example, the OLA platform was integrated with an optofluidic transport assay to create a complete microfluidic total analysis system for quantifying drug permeability. Norfloxacin and ciprofloxacin transport through liposomes was measured at physiological pH and salt concentrations. 138 Vaezi et al produced liposomes to encapsulate taxotere using the OLA. Combined with a microfluidic cell analysis system, encapsulated liposomes' efficacy was analyzed within a single cell in a trapping chip for quantifying drug permeability and apoptosis assay. 139 The EE of taxotere-loaded liposome was 65.49 ± 3.08%, as reported. 139 Niederholtmeyer et al developed porous cellmimics capable of gene expression and communication via diffusive protein signals utilizing OLA. 140 In addition, OLA has proven to be a powerful tool for controlling and studying the formation of membrane-less organelles within liposomes, which help facilitate the bottom-up creation of synthetic cells. 141 First reported by Funakoshi et al, 142 this method mimicked the process of blowing soap bubbles. The pulsed jet flow generated from a micro-nozzle was applied to a planar lipid bilayer formed by the droplet contact method, as the bilayer then stretched and formed free vesicles eventually ( Figure 11A ). 143 The size of the vesicles produced could be tuned by different jet dispensing times. A large amount of monodisperse GUVs (300 μm −600 μm) can be prepared by repeating this process, accompanied by the formation of a small amount of smaller "satellite vesicles". However, residual organic solvent (n-decane) was observed inside the membrane. Subsequently, the same team proved that smaller "satellite vesicles" are generated due to the inhomogeneous break-up of the deformed lipid microtubes. Based on the "satellite vesicles", they prepared asymmetric GUVs with a diameter of 3-20 μm by modifying parameters like the area of the planar lipid bilayer and the jet dispensing time ( Figure 11B ). 144 They confirmed little residual amount of oil in these GUVs using a Raman scattering microscope. Flip-flop motions of lipid molecules in the asymmetric GUVs were observed, indicating that the presence of n-decane between the leaflets had little effect on membrane dynamics. 144 Recently, the team successfully produced cell-sized vesicles containing vesicles, namely MVV, generated by applying pulsed jet flow against two parallel planar asymmetric lipid bilayers formed in a triple-well device, similar to the formation of W/O/W droplets to some extent. 145 Moreover, by applying pulsed-jet flows of longer duration and higher pressure to an asymmetric planar lipid bilayer, nano-sized asymmetric lipid vesicles were formed. The resulting vesicles, however, were polydisperse, and around 30% were multilamellar. 146 Unlike the microdispenser applied by Funakoshi et al, Stachowiak et al used a piezoelectric inkjet nozzle to perform microfluidic jetting. 147, 148 GUVs 10-400 μm in diameter were formed by varying the pulse amplitude, the number of pulses and fluid viscosity. Utilizing this technology, they synthesized asymmetric GUVs with oriented membrane protein incorporation and constructed a synthetic system in which membrane proteins were delivered to the outside of GUVs mimicking aspects of exocytosis. 149 The problem with this method is that the formation of the bilayer produced by manual pipetting requires a certain incubation time, the rate of membrane regrowth limits the vesicle production rate. Also, the difficulty in repositioning the micronozzle relative to the planar lipid bilayer can hinder repeatability and throughput. As a result, fast and continuous preparation of liposomes could not be prepared in large quantities via this method. The piezoelectric inkjet technology could increase the production rate by up to 200 Hz in a way, but a pause of 2-4 seconds was needed for the lipid bilayer to regrow. 148 To address this problem, Gotanda et al arranged the six wells on a circular rotation platform to prepare various asymmetric liposomes sequentially by accurately controlling rotation speed parameters and jetting timing through an automated system. Approximately 300 asymmetric cell-sized GUVs (mean diameter of 8.9 ± 6.7 μm) were collected within 2 minutes. 150 Armstrong et al pioneered the use of focused high-intensity ultrasound instead of micro-nozzles to induce focused flow jets to prepare GUVs ( Figure 11C ), eliminating stability challenges. GUVs of 100-300 μm in diameter were formed by controlling the numerical aperture of the acoustic lens and the acoustic wavelength. 151 Notably, ultrasound itself will affect the stability of liposomes. It is crucial to ensure the stability of planar lipid bilayers during the formation of vesicles. In this method, materials such as DNA, cells, and so on, regardless of their size, concentration, or chemical properties, can be directly encapsulated into liposomes by jetting, similar to pico injection, bringing about a high EE (~90%) as reported. 144 However, organic solvent remnants were found between leaflets in this method. 142, 144, 152 Thus, sophisticated equipment demanding operating conditions with low liposome yield confines its further applications other than membrane science and synthetic cells. Nevertheless Thin-film hydration is a common method used for the formation of vesicles. The process involves depositing lipid films onto a substrate after evaporation of the organic solvent. The films then swell, peel off, and eventually selfassemble into liposomes in the absence of an aqueous solution. Since there is no control over the hydration process, most of the liposomes prepared are polydisperse MLVs. Subsequent extrusion or other treatments are required to obtain smaller monodisperse UVs. [153] [154] [155] To handle this problem, Osaki et al focused on the control of lipid deposition process. Arrayed micro apertures were fabricated onto an indium tin oxide (ITO)-glass slide by soft lithography. Following the electrospray deposition (ESD) method, selectively patterned lipid films were formed on the conductive surface ( Figure 12 ). 156 With a simple hydration process of the dried lipid, they succeeded in forming giant liposomes on top of the pattern. Although ESD could prepare monodisperse giant vesicles, the area of the sprayed films is too small at once to obtain a high yield. One drawback of hydration is the low EE as vesicles cannot be isolated from materials in the external environment during the formation process. 157 Moreover, the process requires the deposition of a lipid film on a substrate followed by time-consuming evaporation, which prevents the continuous preparation of vesicles since lipids cannot be replenished during operation. Other microfluidic methods for preparing liposomes, such as micromixers, are more practical when applied to drug delivery. The electroformation method for the production of giant liposomes was first described by Angelova and Dimitrov in 1986. 30 Similar to hydration, dried lipid films were prepared on the surface of electrodes. An electric field was then applied to the hydrated films immersed in an aqueous solution, resulting in the formation of giant vesicles. However, the electroformation method has a low yield of monodisperse vesicles and lacks an efficient collection process of prepared liposomes. 30, 154, 158 Kuribayashi et al succeeded in integrating the electroformation method within microfluidic chips. 159 Two parallel microfluidic channels formed in the silicone sheet were sandwiched between the ITO glass plates to prepare giant vesicles. Liposomes with different lipid compositions prepared by this method could encapsulate different types of materials. Since they were polydisperse, microcontact printing was applied to control the size distribution of vesicles. [160] [161] [162] The lipid is deposited 163, 164 Recently, Wang et al integrated the preparation, separation, and collection of liposomes onto a PDMS chip. After electroformation, liposomes were driven by fresh buffer and trapped by filter membranes in a tailor-made collection chamber, which increased the liposome collection ratio to 40%. 165 In 2013, Bi et al introduced the use of coplanarinterdigitated electrodes for electroformation of liposomes ( Figure 13 ), breaking from the tradition of using two opposite electrodes. 166 Smaller interdigitated electrodes generate bigger GUVs in this method, which provides a reliable solution for controlling the size distribution of giant liposomes by electroformation. Apart from those defects discussed in the hydration method, irregular and asymmetric lipid distribution was observed in GUVs formed by electroformation. 167 The electric field itself may have adverse effects on liposomes described in Micromixer. 98, 99 Recent research has mostly focused on exploring novel microfluidics-based electroformation methods for liposomes preparation, and few related applications have been reported, 167, 168 to some extent implying that applications of electroforming methods are limited due to the shortcomings described above. With the successful development of micro-and nanofabrication, 169 the preparation of liposomes has been transposed from the macro-to the micro-scale. Microfluidic technologies can not only precisely control the size and lamellarity of liposomes but also reduce the volume and cost of reagents required simultaneously. Comparison between various microfluidic methods used to prepare liposomes is briefly summarized in Table 2 . Different methods have their pros and cons. Thin-film hydration and electroformation are more biocompatible, avoiding the presence of organic solvents. The issue of low EE is insignificant when applied to membrane science. Although the pulse flow jetting and single emulsion templates with higher EE are more difficult to implement, they are near-perfect techniques for synthetic cells. Double emulsion templates, especially the OLA method, are also useful tools to prepare GUVs to explore new drugs and elucidate cellular functions. For the large-scale production of liposomes/LNPs for drug delivery, micromixers may be preferred due to their high throughput. In the case of single emulsion templates, combining dsGUVs with pico injection is one of the few means of directly encapsulating any desired materials into liposomes with the assistance of pico injection technology, which circumvents dilution or contamination of encapsulated materials. Obviously, LNPs are not suitable for larger macromolecules with radii of gyration as large as 1 μm, which may be required for potential applications, such as drug delivery and gene therapy. Moreover, their various strategies for biofunctionalization of GUVs (charged, bioligand-conjugated or PEG-conjugated) widened the horizons in applications of GUVs, such as targeted cellular delivery. Potentially immunogenic cargo could be intracellularly discharged via the lysosomal escape mechanism once the carriers arrive at their destination without initiating an immune response. Work by Spatz group presented an exquisite model for combining liposomes and microfluidic techniques, 107,109 motivating further fundamental and applied liposomes research. Coincidentally, several studies have been utilizing other external energies such as acoustic waves and electromagnetics to boost current microfluidic methods by enhancing production efficiency 96, 97 or simplifying the manufacture of equipment. 151 These initiatives have supported the recent efforts to investigate and improve nanotechnology for liposomes formation, but future investigations must critically evaluate whether the properties of liposomes are prone to be disrupted by physical methods like ultrasound and electric field. No matter which method, bulk or microfluidic, it is necessary to separate and purify liposomes after preparation before further applications. Some microfluidic methods, such as single and double emulsion templates, may require additional post-processing steps ( Table 2) . For example, the floating oil droplets formed by quick dewetting of the double-emulsion droplets had to be removed by an extra step of pipetting or evaporating. 137 OLA also suffered from the presence of 1-octanol droplets, an inevitable by-product of the production process. 135 To separate the 1-octanol droplets from the liposomes formed by OLA, a one-step density-based separation technique integrated on the same chip had been developed by the same group. 170 A high separation efficiency (~93%) was realized compared to the non-separated samples (~45%). Similarly, Dimov et al introduced a microfluidic system composed of SHM and a tangential flow filtration device for consecutive purification. This on-chip platform can eliminate most non-encapsulated materials and organic solvent (>90%) in less than 4 minutes, offering simultaneous production and purification of liposomes. 171 Although purification steps (described in Encapsulation Efficiency) could be conducted off-chip, retaining the liposomes inside the microfluidic chip is of great significance. It reduces purification time, eliminates the possible loss of material during off-chip handling, and, more importantly, enables downstream on-chip experimentation. In fact, endeavors have been made to establish one-step microfluidic platforms for subsequent observation, purification or manipulation of liposomes, such as capture and immobilization, 116, 172 separations and purification, 165 dynamic monitoring 173 and even functional evaluation. 138, 174 These on-chip efforts will certainly broaden the horizons for the research of liposomes/LNPs. LNPs, an upgraded version of liposomes, have now been utilized as a versatile nanomedicine delivery platform. Although LNPs are currently in the spotlight because of the success of the COVID-19 mRNA vaccines, viral vectors are more frequently used than LNPs since they are more efficient in delivering DNA. RNA only needs to enter the cytoplasm to function, while gene therapy demands the delivery of DNA into the nucleus through the nuclear membrane. Effectively delivering DNA molecules in vitro and in vivo using LNPs requires more in-depth investigations of lipid nanoparticle-DNA formulations. Most liposomes/LNPs are undoubtedly used in medical science, but academics who usually use these nanoparticles for drug delivery prefer conventional bulk methods, such as ethanol dilution, solvent injection or homogenization to prepare liposomes. 19, 175, 176 They concentrate on designing and optimizing the nanocarrier itself instead of novel techniques for nanocarrier preparation. Since the production techniques cannot resolve the critical clinical problems in liposomal applications, such as delivery efficacy, organ-specific and cell-specific delivery and toxicity, to mention a few, it was not surprising that these bulk methods were chosen due to simplicity, costsaving and safety. However, researchers that promote the development of preparation techniques pay more attention to the basic design of microfluidic devices like the geometry or the fundamental physics behind the microfluidics-assisted nanoparticle productions, such as the dynamics of the process. 177 The blossoming of liposomes/LNPs requires the synergistic development of miscellaneous fields, including medical, physics, chemistry and engineering. In light of this review, we encourage researchers to not only choose the most suitable techniques for the desired research but also bravely explore possible applications of newer techniques. This will likely change in the near future as the popularity of commercial micromixers (eg, the NanoAssemblr™ platform) increased. 178, 179 Based on the current progress and success, we firmly believe that microfluidics will soon revolutionize the generation of sophisticated artificial cells and lipid-based nanoencapsulation. work was supported by the National Key R&D Program of China (2019YFA0110500) and the National Natural Science Foundation of China (No. 8201001114). Negative staining of phospholipids and their structural modification by surface-active agents as observed in the electron microscope Diffusion of univalent ions across the lamellae of swollen phospholipids Liposomes loaded with a STING pathway ligand, cyclic di-GMP, enhance cancer immunotherapy against metastatic melanoma Design of lipid nanoparticles for in vitro and in vivo delivery of plasmid DNA Tumor microenvironment activated membrane fusogenic liposome with speedy antibody and doxorubicin delivery for synergistic treatment of metastatic tumors Cationic liposomes carrying siRNA: impact of lipid composition on physicochemical properties, cytotoxicity and endosomal escape Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathways Cholesterol modulates the liposome membrane fluidity and permeability for a hydrophilic molecule Peptide-mediated liposome fusion: the effect of anchor positioning Protein synthesis in artificial cells: using compartmentalisation for spatial organisation in vesicle bioreactors A DNA-programmed liposome fusion cascade Self-replication of DNA by its encoded proteins in liposome-based synthetic cells Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine Role of nanotechnology behind the success of mRNA vaccines for COVID-19 Updated survival analysis in patients with stage IIIB or IV non-small-cell lung cancer receiving BLP25 liposome vaccine (L-BLP25): phase IIB randomized, multicenter, open-label trial Tumortargeting dual peptides-modified cationic liposomes for delivery of siRNA and docetaxel to gliomas A single-dose mRNA vaccine provides a long-term protection for hACE2 transgenic mice from SARS-CoV-2 SARS-CoV-2 RBD neutralizing antibody induction is enhanced by particulate vaccination Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing Membrane-destabilizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing Microfluidic assembly of monodisperse vesosomes as artificial cell models Microfluidic formation of monodisperse coacervate organelles in liposomes Controlled membrane translocation provides a mechanism for signal transduction and amplification Preparation of liposomes using an improved supercritical reverse phase evaporation method Ethanol injection technique for liposomes formulation: an insight into development, influencing factors, challenges and applications Gene expression within cell-sized lipid vesicles Structure of fully hydrated fluid phase DMPC and DLPC lipid bilayers using X-ray scattering from oriented multilamellar arrays and from unilamellar vesicles Filter extrusion of liposomes using different devices: comparison of liposome size, encapsulation efficiency, and process characteristics Characterization of liposomes. The influence of extrusion of multilamellar vesicles through polycarbonate membranes on particle size, particle size distribution and number of bilayers Liposome electroformation Electroformation of giant liposomes from spin-coated films of lipids A novel method for the preparation of liposomes: freeze drying of monophase solutions Freeze-drying of liposomes using tertiary butyl alcohol/water cosolvent systems Engineering asymmetric vesicles A lipid based multi-compartmental system: liposomes-in-double emulsion for oral vaccine delivery The size of liposomes: a factor which affects their targeting efficiency to tumors and therapeutic activity of liposomal antitumor drugs Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size Liposomal drug delivery systems: from concept to clinical applications Microfluidic methods for forming liposomes Liposome production by microfluidics: potential and limiting factors A comprehensive review on recent preparation techniques of liposomes Liposome: classification, preparation, and applications Critical evaluation of nanoparticle tracking analysis (NTA) by nanosight for the measurement of nanoparticles and protein aggregates Engineering asymmetric lipid vesicles: accurate and convenient control of the outer leaflet lipid composition Thermal and 31P-NMR studies to elucidate sumatriptan succinate entrapment behavior in phosphatidylcholine/cholesterol liposomes. Comparative 31P-NMR analysis on negatively and positively-charged liposomes Microfluidic platform for the continuous production and characterization of multilamellar vesicles: a synchrotron small-angle X-ray scattering (SAXS) study In-situ transmission electron microscopy of liposomes in an aqueous environment Extended pain relief trial utilizing infiltration of Exparel( ® ), a long-acting multivesicular liposome formulation of bupivacaine: a phase IV health economic trial in adult patients undergoing open colectomy Formulation and evaluation of multilamellar vesicles ropivacaine in pain management Role of liposome size and RES blockade in controlling biodistribution and tumor uptake of GM1-containing liposomes Opportunities and challenges in commercial pharmaceutical liposome applications Microfluidic handling and analysis of giant vesicles for use as artificial cells: a review Influence of cholesterol on liposome stability and on in vitro drug release A pH-sensitive cationic lipid facilitates the delivery of liposomal siRNA and gene silencing activity in vitro and in vivo Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses Synthesis and characterization of PEGylated bolaamphiphiles with enhanced retention in liposomes biocompatibility and antioxidant activity of PEG-modified liposomes containing resveratrol Soft interaction in liposome nanocarriers for therapeutic drug delivery Effect of charge on separation of liposomes upon stagnation Zeta potential measurement. In: Characterization of Nanoparticles Intended for Drug Delivery Liposomal nanovesicles for efficient encapsulation of staphylococcal antibiotics Liposome retention in size exclusion chromatography A study on characteristic of different sample pretreatment methods to evaluate the entrapment efficiency of liposomes Determination of liposomal encapsulation efficiency using proton NMR spectroscopy Encapsulation efficiency measured on single small unilamellar vesicles Determination of the encapsulation efficiency of individual vesicles using single-vesicle photolysis and confocal single-molecule detection A simple and rapid measurement method of encapsulation efficiency of doxorubicin loaded liposomes by direct injection of the liposomal suspension to liquid chromatography Controlled vesicle self-assembly in microfluidic channels with hydrodynamic focusing Microfluidic directed formation of liposomes of controlled size Microfluidic mixing and the formation of nanoscale lipid vesicles Rapid, one-step fabrication and loading of nanoscale 1,2-distearoyl-sn-glycero-3-phosphocholine liposomes in a simple, double flow-focusing microfluidic device Microfluidic hydrodynamic focusing based synthesis of POPC liposomes for model biological systems Continuous-flow production of injectable liposomes via a microfluidic approach Freezing continuous-flow self-assembly in a microfluidic device: toward imaging of liposome formation Continuous flow production of cationic liposomes at high lipid concentration in microfluidic devices for gene delivery applications Continuous production of the nanoscale liposome in a double flow-focusing microfluidic device A facile route to the synthesis of monodisperse nanoscale liposomes using 3D microfluidic hydrodynamic focusing in a concentric capillary array High-throughput continuous flow production of nanoscale liposomes by microfluidic vertical flow focusing High-throughput continuous production of liposomes using hydrodynamic flow-focusing microfluidic devices High throughput nanoliposome formation using 3D printed microfluidic flow focusing chips Microfluidic devices for continuous production of pDNA/cationic liposome complexes for gene delivery and vaccine therapy Microfluidic synthesis of multifunctional liposomes for tumour targeting Double flow focusing microfluidic-assisted based preparation of Methotrexate-Loaded liposomal nanoparticles: encapsulation efficacy, drug release and stability Microfluidic remote loading for rapid single-step liposomal drug preparation Remote loading of preencapsulated drugs into stealth liposomes Chaotic mixer for microchannels Bottom-up design and synthesis of limit size lipid nanoparticle systems with aqueous and triglyceride cores using millisecond microfluidic mixing High-throughput manufacturing of size-tuned liposomes by a new microfluidics method using enhanced statistical tools for characterization A strategy for synthesis of lipid nanoparticles using microfluidic devices with a mixer structure A novel microfluidic-based approach to formulate size-tuneable large unilamellar cationic liposomes: formulation, cellular uptake and biodistribution investigations Understanding the formation mechanism of lipid nanoparticles in microfluidic devices with chaotic micromixers Microfluidics based manufacture of liposomes simultaneously entrapping hydrophilic and lipophilic drugs Lipid nanoparticlesfrom liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement Engineering precision nanoparticles for drug delivery Development of the iLiNP device: fine tuning the lipid nanoparticle size within 10 nm for drug delivery An ultra-rapid acoustic micromixer for synthesis of organic nanoparticles Electrohydrodynamic-driven micromixing for the synthesis of highly monodisperse nanoscale liposomes Optimization of the electroformation of giant unilamellar vesicles (GUVs) with unsaturated phospholipids Low levels of lipid oxidation radically increase the passive permeability of lipid bilayers Microfluidic-controlled manufacture of liposomes for the solubilisation of a poorly water soluble drug Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA Lipid nanoparticles containing siRNA synthesized by microfluidic mixing exhibit an electron-dense nanostructured core Scalable mRNA and siRNA lipid nanoparticle production using a parallelized microfluidic device CRISPR-Cas9 genome editing using targeted lipid nanoparticles for cancer therapy Ionizable lipid nanoparticles for in utero mRNA delivery Sequential bottom-up assembly of mechanically stabilized synthetic cells by microfluidics Highthroughput injection with microfluidics using picoinjectors Microfluidic production and characterization of biofunctionalized giant unilamellar vesicles for targeted intracellular cargo delivery Novel method for obtaining homogeneous giant vesicles from a monodisperse water-in-oil emulsion prepared with a microfluidic device Efficient preparation of giant vesicles as biomimetic compartment systems with high entrapment yields for biomacromolecules Production of unilamellar vesicles using an inverted emulsion Controlled microfluidic encapsulation of cells, proteins, and microbeads in lipid vesicles Microfluidic fabrication of asymmetric giant lipid vesicles Size control of giant unilamellar vesicles prepared from inverted emulsion droplets Stepwise synthesis of giant unilamellar vesicles on a microfluidic assembly line Preparation and mechanical characterisation of giant unilamellar vesicles by a microfluidic method Studying the effects of asymmetry on the bending rigidity of lipid membranes formed by microfluidics Layer-by-layer cell membrane assembly Droplet-shooting and size-filtration (DSSF) method for synthesis of cell-sized liposomes with controlled lipid compositions Direct observation of bacterial growth in giant unilamellar vesicles: a novel tool for bacterial cultures. ChemistryOpen Preparing protein producing synthetic cells using cell free bacterial extracts, liposomes and emulsion transfer Characterization of giant vesicles formed by phase transfer processes Double emulsion templated monodisperse phospholipid vesicles Double-emulsion drops with ultra-thin shells for capsule templates Ultrathin shell double emulsion templated giant unilamellar lipid vesicles with controlled microdomain formation Cell-free compartmentalized protein synthesis inside double emulsion templated liposomes with in vitro synthesized and assembled ribosomes One-step microfluidics production of enzyme-loaded liposomes for the treatment of inflammatory diseases Stable, biocompatible lipid vesicle generation by solvent extraction-based droplet microfluidics Vesicles-on-a-chip: a universal microfluidic platform for the assembly of liposomes and polymersomes Membrane mechanical properties of synthetic asymmetric phospholipid vesicles Single-step assembly of asymmetric vesicles Continuous microfluidic fabrication of synthetic asymmetric vesicles Solvent compatibility of poly(dimethylsiloxane)-based microfluidic devices Octanol-assisted liposome assembly on chip On-chip microfluidic production of cell-sized liposomes Monodisperse uni-and multicompartment liposomes An integrated microfluidic platform for quantifying drug permeation across biomimetic vesicle membranes Investigation of the programmed cell death by encapsulated cytoskeleton drug liposomes using a microfluidic platform Communication and quorum sensing in non-living mimics of eukaryotic cells Spatiotemporal control of coacervate formation within liposomes Formation of giant lipid vesicle like compartments from a planar lipid membrane by a pulsed jet flow Lipid bilayer formation by contacting monolayers in a microfluidic device for membrane protein analysis Cellsized asymmetric lipid vesicles facilitate the investigation of asymmetric membranes Formation of vesicles-in-a-vesicle with asymmetric lipid components using a pulsed-jet flow method Formation of nano-sized lipid vesicles with asymmetric lipid components using a pulsed-jet flow method Unilamellar vesicle formation and encapsulation by microfluidic jetting Inkjet formation of unilamellar lipid vesicles for cell-like encapsulation Forming giant vesicles with controlled membrane composition, asymmetry, and contents Automatic generation system of cell-sized liposomes Forming and loading giant unilamellar vesicles with acoustic jetting Membrane composition of jetted lipid vesicles: a Raman spectroscopy study Formation and properties of thin-walled phospholipid vesicles Giant vesicles formed by gentle hydration and electroformation: a comparison by fluorescence microscopy Thin-film hydration followed by extrusion method for liposome preparation Uniformly-sized giant liposome formation with gentle hydration A novel method for the encapsulation of biomolecules into polymersomes via direct hydration Efficient electroformation of supergiant unilamellar vesicles containing cationic lipids on ITO-coated electrodes Electroformation of giant liposomes in microfluidic channels A novel technique for preparation of monodisperse giant liposomes Fabrication of 2D arrays of giant liposomes on solid substrates by microcontact printing Multifunctional lipid multilayer stamping Electroformation of giant phospholipid vesicles on a silicon substrate: advantages of controllable surface properties Preparation of phospholipid multilayer patterns of controlled size and thickness by capillary assembly on a microstructured substrate Electroformation and collection of giant liposomes on an integrated microchip Electroformation of giant unilamellar vesicles using interdigitated ITO electrodes Charged giant unilamellar vesicles prepared by electroformation exhibit nanotubes and transbilayer lipid asymmetry A convenient protocol for generating giant unilamellar vesicles containing SNARE proteins using electroformation Hydration-enhanced lubricating electrospun nanofibrous membranes prevent tissue adhesion On-chip density-based purification of liposomes Formation and purification of tailored liposomes for drug delivery using a module-based micro continuous-flow system Charge-controlled microfluidic formation of lipid-based single-and multicompartment systems Rapid liposome quality assessment using a lab-on-a-chip A microfluidic tumor-on-a-chip for assessing multifunctional liposomes' tumor targeting and anticancer efficacy Theranostic liposomes of TPGS coating for targeted co-delivery of docetaxel and quantum dots Influence of lipid matrix composition on biopharmaceutical properties of lipid nanoparticles Microfluidics: microfluidics for production of particles: mechanism, methodology, and applications (Small 9/2020) Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves liver-specific in vivo genome editing of Angptl3 Design of SARS-CoV-2 hFc-conjugated receptor-binding domain mRNA vaccine delivered via lipid nanoparticles and the Elsevier Bibliographic databases. The manuscript management system is completely online and includes a very quick and fair peer-review system The authors thank Dr. Zhenxing Wang for his constructive comments during the preparation of this manuscript. This The authors have no conflicts of interest to declare that are relevant to the content of this article.