Droplet microfluidics has emerged as a versatile and powerful tool for various analytical applications, including single-cell studies, synthetic biology, directed evolution, and diagnostics. Initially, access to droplet microfluidics was predominantly limited to specialized technology labs. However, the landscape is shifting with the increasing availability of commercialized droplet manipulation technologies, thereby expanding its use to non-specialized labs. Although these commercial solutions offer robust platforms, their adaptability is often constrained compared to in-house developed devices. Consequently, both within the industry and academia, significant efforts are being made to further enhance the robustness and automation of droplet-based platforms, not only to facilitate technology transfer to non-expert laboratories but also to reduce experimental failures. This Perspective article provides an overview of recent advancements aimed at increasing the robustness and accessibility of systems enabling complex droplet manipulations. The discussion encompasses diverse aspects such as droplet generation, reagent addition, splitting, washing, incubation, sorting, and dispensing. Moreover, alternative techniques like double emulsions and hydrogel capsules, minimizing or eliminating the need for microfluidic operations by the end user, are explored. These developments are foreseen to facilitate the integration of intricate droplet manipulations by non-expert users in their workflows, thereby fostering broader and faster adoption across scientific domains.

Droplet-based microfluidics has emerged as a versatile and powerful tool for a wide range of high-impact applications, such as single-cell analysis,1–4 high-throughput screening,5–7 synthetic biology,8,9 precision medicine,10–12 and diagnostics.13–15 In the decade following the first report of water-in-oil (w/o) droplets formed by microfluidics in 1997,16 most endeavors focused on the development of methods for high-throughput monodisperse droplet generation,17,18 sample encapsulation,19 and droplet manipulations such as splitting,20 reagent addition,21 incubation,22 and sorting.17,23 However, at that time, the adoption of droplet microfluidics was largely confined to specialized technology labs, limiting its accessibility to researchers in other fields who could in fact benefit most from the technology. Collaboration with experts in microfluidics was often a prerequisite for utilizing this transformative technology effectively. In recent years, the landscape of droplet microfluidics has undergone a remarkable transformation. Although droplet manipulation technologies are continuously being developed and improved, the paradigm has shifted toward the implementation of groundbreaking biological or chemical assays inside the w/o droplets.

Driven by the requirements posed by scientists to translate their assays to droplet microfluidics, three noticeable developments to improve accessibility of droplet microfluidic workflows have emerged, which will be discussed in this Perspective article. First of all, increasing efforts are made on advancing robustness and automation of w/o droplet microfluidics, as well as on implementing multiple, complex manipulations in a single workflow to enable applications with highly demanding prerequisites. These efforts can be found both in academic literature as well as in the commercial field. Second, researchers in various scientific domains are gaining unprecedented access to w/o droplet microfluidic platforms, thanks to the commercialization of several droplet manipulation systems. These commercial systems have varying levels of automation and flexibility. Finally, alternatives to w/o droplets are arising, such as double emulsions and hydrogel capsules, which attempt to offer more user-friendly workflows by integrating standard laboratory equipment, while maintaining advantages of w/o droplet systems. We conclude the article with a future outlook, shedding light on the promising future of droplet microfluidics as a widely accessible and versatile tool in various scientific disciplines.

Thus, the aim of this article is to give a perspective on how the field of droplet microfluidics is shifting from a specialized domain toward broad adoption, fueled by advancements that improve the robustness and automation of droplet manipulations, along with the increasing availability of commercially accessible systems and alternatives to traditional w/o droplets. Unlike other reviews, this article provides only a brief description of the common droplet manipulations, focusing primarily on recent developments aimed at enhancing reliability and usability. The article serves a dual purpose: first, to inform non-experts about the potential and challenges of droplet microfluidics and to offer guidance on selecting the most suitable and accessible technologies for their needs. Second, it provides experts with insights into emerging trends in robust droplet manipulations, encouraging them to design solutions that prioritize end-user accessibility and practical application.

Since this article primarily focuses on droplet manipulation techniques, we refer the reader to other literature for in-depth discussions on chip fabrication techniques,24,25 sample encapsulation in droplets,26,27 and droplet read-out systems.28,2 Moreover, this article focuses on channel-based droplet microfluidics, thereby omitting digital microfluidic systems using (optical) electro-wetting-on-dielectric30,31 or droplet-digital microfluidic hybrid systems.32–34 

The primary step in all droplet microfluidic systems is droplet generation, during which w/o emulsions are conventionally generated by passive methods (e.g., via T-junctions or flow-focusing structures).35 Droplets with diameters of a few to hundreds of μm (corresponding to fl to nl volumes) can be generated at hundreds to ten-thousands per second, where the generation throughput is typically inversely related to the droplet size. Key factors to take into account during droplet generation are (1) the required droplet size, which is influenced by the microfluidic geometry, (2) the operational flow rates, and (3) the liquid properties (interfacial tension and viscosity). However, for given liquid properties and a particular microfluidic droplet generation chip, the user can tune the droplet dimensions only to a limited extent by adjusting flow rates. Hence, proper chip selection is required, which might be cumbersome for the non-expert user. Therefore, recent research focusses on developing a framework that enables researchers to make informed decisions on the channel dimensions and flow conditions, required to obtain droplets of the size of interest.36 In this context, it is also of paramount importance that the generated droplets are monodisperse, for which a highly accurate flow control system is required (e.g., syringe- or pressure-based pumps). Although these systems typically enable generation of droplets with coefficients of variation between 2% and 5%, during prolonged experiments the droplet sizes can drift for a variety of reasons. To render the droplet generation process more robust, recent efforts focus on including image-based closed-loop feedback systems during droplet generation (Fig. 1), in which in-line droplet size monitoring and coupled adjustments to the pressures or flow rates ensure drastic improvement of monodispersity over prolonged running times.37–39 This is exemplified by Crawford et al. who showed an almost 10-fold decrease (from 3.8% to 0.4%) in coefficient of variation (CV) in droplet volume between pressure pump-based and feedback system-based droplet generation over 45 min.38 

FIG. 1.

Image-based closed-loop feedback system for robust droplet generation, where the droplet size is detected in real time during generation and the aqueous and oil flow rates or pressures are adjusted accordingly to ensure generation of monodisperse droplets over prolonged times. P = pressure, Q = flow rate, bf = bright field. Adapted with permission from Crawford et al. Sci. Rep. 7(1), 10545 (2017). Copyright 2017 Author(s), licensed under a Creative Commons Attribution 4.0 License.

FIG. 1.

Image-based closed-loop feedback system for robust droplet generation, where the droplet size is detected in real time during generation and the aqueous and oil flow rates or pressures are adjusted accordingly to ensure generation of monodisperse droplets over prolonged times. P = pressure, Q = flow rate, bf = bright field. Adapted with permission from Crawford et al. Sci. Rep. 7(1), 10545 (2017). Copyright 2017 Author(s), licensed under a Creative Commons Attribution 4.0 License.

Close modal

The most commonly used methods for reagent addition into w/o droplets are droplet merging and picoinjection. In droplet merging, two or more sequences of droplets, either formed on-chip or pre-formed and reintroduced, are paired and fused, with fusion occurring either actively under the influence of, e.g., an acoustic40 or electric field41–43 or passively due to hydrodynamic forces44–46 at rates of up to several kHz. Various droplet merging techniques, encompassing single and combinatorial droplet merging, have been explored, but precise synchronization between different droplet sequences remains the key challenge in order to achieve robust one-on-one droplet pairing. As a result, the user typically needs to continuously observe the experiment and adjust flow parameters in order to maintain accurate droplet pairing. In this context, developments in automated, passive droplet pairing based on self-synchronization through precise channel designs are emerging, reaching over 99% one-on-one pairing efficiency at a rate of 111 (Ref. 42) and 200 Hz,47 as compared to up to 95% efficiency by systems not implementing this design47 (Fig. 2).

FIG. 2.

Hydrodynamic resistance-regulated device for improved synchronization and merging of two droplet sets. Synchronization is achieved by transient blocking of subsequent droplets to enter the channel by the increased hydrodynamic resistance, originating from a droplet inside the constriction site. Adapted with permission from Nan et al., Microsyst. Nanoeng. 9(1), 24 (2023). Copyright 2023 Author(s), licensed under a Creative Commons Attribution 4.0 License.

FIG. 2.

Hydrodynamic resistance-regulated device for improved synchronization and merging of two droplet sets. Synchronization is achieved by transient blocking of subsequent droplets to enter the channel by the increased hydrodynamic resistance, originating from a droplet inside the constriction site. Adapted with permission from Nan et al., Microsyst. Nanoeng. 9(1), 24 (2023). Copyright 2023 Author(s), licensed under a Creative Commons Attribution 4.0 License.

Close modal

Picoinjection involves reagent addition through pressurized liquid in a side-channel that merges with passing droplets in the main channel under the influence of, e.g., an electric48–50 or acoustic51 field and has been reported up to 10 kHz.48 This method can employ either a single injector or multiple serial injectors for combinatorial reagent addition. The first-reported picoinjection approach utilized pressure to set the injector at the Laplace pressure, i.e., the pressure at which the water in the injector channel and the oil in the main channel are at a stable equilibrium, with an electric field triggering the injection.48 However, due to pressure fluctuations, injection using the equilibrium pressure is not robust for longer experiment times, leading to failed injection, drift in injection volume, or the generation of droplets by the injector. As such, similarly, as for merging, the user needs to observe the experiment continuously to make adjustments to flow parameters. Therefore, over time, improvements have been sought, such as including the implementation of pressure stabilizers which enable a 10-fold reduction in internal pressure at the picoinjector during fluctuations.49 Additionally, syringe pumps to control the injected volume by varying flow rates52–55 can be used to fully eliminate the need to set the equilibrium pressure. While injection by flow rate control is robust and even allows for electrode-free systems,56–58 precisely turning off picoinjection is challenging. A semi-automated method for combined flow rate and pressure control of up to three serial picoinjectors has recently emerged (Fig. 3), allowing them to inject varying volumes as well as to turn off injection, respectively, in an unsupervised way. This is achieved by performing a calibration of the equilibrium pressure, which is then automatically adjusted in real-time.50 However, the pressure calibration needs to be performed manually and requires significant microfluidics expertise. Recently, triggered picoinjection59–61 and droplet merging62 have also been reported, where reagents are added only to droplets of interest, allowing to truly control the content of individual droplets. For both droplet merging and picoinjection, implementing camera-based feedback, as previously performed for low throughput droplet manipulations,63 could enhance the robustness and automation of these methods.22 

FIG. 3.

Combined flow rate and pressure control for robust serial picoinjection, through calibration of the system and continuous adjustment of the Laplace pressure. A linear relationship between the Laplace pressure of the injector and the oil pressure is defined, which is then used to adjust the Laplace pressure in real-time to accurately turn off picoinjection. During picoinjection, flow rate control is used, where the applied pressure is continuously adjusted to reach the required flow rate as measured by flow sensors. P = pressure, Q = flow rate. Adapted with permission from Breukers et al., Lab Chip 22(18), 3475–3488 (2022). Copyright 2022 Royal Society of Chemistry.

FIG. 3.

Combined flow rate and pressure control for robust serial picoinjection, through calibration of the system and continuous adjustment of the Laplace pressure. A linear relationship between the Laplace pressure of the injector and the oil pressure is defined, which is then used to adjust the Laplace pressure in real-time to accurately turn off picoinjection. During picoinjection, flow rate control is used, where the applied pressure is continuously adjusted to reach the required flow rate as measured by flow sensors. P = pressure, Q = flow rate. Adapted with permission from Breukers et al., Lab Chip 22(18), 3475–3488 (2022). Copyright 2022 Royal Society of Chemistry.

Close modal

Droplets can be split into two or more daughter droplets of equal or distinct sizes. Several active mechanisms have been described for splitting, including mechanical64,65 and acoustic66 systems. However, thanks to its simplicity, most splitting systems rely on passive mechanisms, by utilizing approaches such as standard flow-focusing geometries,67,68 akin to those employed in droplet generation, or a combination of specific channel geometries (e.g., bifurcations) and hydrodynamic forces.20,69–72 Passive droplet splitting systems, especially when carefully designing the microchannels, hold particular significance for robust automation and can operate at speeds of several kHz. One major challenge in the application of droplet splitting is the ability to link different daughter droplets, which can be of interest when there is a need to couple various readouts to a single mother droplet. In addressing this challenge, accurately designed delay lines (Fig. 4, see also Incubation section) have proven successful to implement readout on one daughter droplet and perform sorting on the other daughter droplet for downstream processing.73,74

FIG. 4.

An example of passive droplet splitting relying on a T-junction. Droplet splitting can be useful to enable processing of droplets in multiple ways that are incompatible with each other. A design with incorporated delay line was used to study the content of one daughter droplet, and, based on the result, perform droplet sorting and downstream processing on the corresponding second daughter droplet.73,74 P = pressure, Q = flow rate. Adapted with permission from Holland-Moritz et al., Angew. Chem. 132(11), 4500–4507 (2020). Copyright 2020 Wiley.

FIG. 4.

An example of passive droplet splitting relying on a T-junction. Droplet splitting can be useful to enable processing of droplets in multiple ways that are incompatible with each other. A design with incorporated delay line was used to study the content of one daughter droplet, and, based on the result, perform droplet sorting and downstream processing on the corresponding second daughter droplet.73,74 P = pressure, Q = flow rate. Adapted with permission from Holland-Moritz et al., Angew. Chem. 132(11), 4500–4507 (2020). Copyright 2020 Wiley.

Close modal

Washing is a relatively underexplored droplet manipulation, primarily due to its inherent complexity. Namely, droplet washing requires alternating between droplet splitting, during which the content of interest is retained in one of the daughter droplets, and reagent addition to the daughter droplet with the content of interest, using a combination of the splitting and addition techniques as described above.58,75–79 To specifically retain cells or microparticles with immobilized molecules into a specific daughter droplet during splitting, either passive mechanical restrictions80 or active forces, such as acoustophoresis,78,79 dielectrophoresis,76 and magnetism58,75,77,81,82 are used. Given the complexity of the workflows, most droplet washing protocols operate at only several Hz, with a few exceptions operating at 500 Hz81 or several kHz.80 Although automating individual steps involved in washing should be feasible, the efficiency of retaining cells and microparticles will depend upon the sample. Additionally, the use of these washing techniques is closely linked to the concurrent development of microparticle-based bioassays. Altogether, these factors may present challenges in the creation of robust, user-friendly devices. Nevertheless, given the critical role of washing in many assays, we anticipate further developments in this area.

In some applications, droplet incubation is crucial to allow (bio)chemical reactions to occur inside the droplets. In this context, some microfluidic systems employ delay lines, featuring long, broad channels where droplets remain in continuous flow. Clever channel design can even maintain the sequential order of droplets, resulting in precise and known incubation times per droplet.22 While this approach is robust, it is only applicable to shorter incubation periods, typically from a few seconds up to 1 h.83 When dealing with large amounts of droplets and extended incubation periods, a common approach is to collect the droplets within reservoirs (e.g., tubing, tubes, and syringes) located off the microfluidic chip.25 After the necessary incubation period, these droplets are subsequently reintroduced into another chip for further analysis or manipulation. However, this off-chip method can be challenging to ensure robustness and automation as it often requires manual interventions, which lead to significant droplet loss and increased chance of spontaneous droplet coalescence. Therefore, to enable on-chip monitoring, arrays designed to trap droplets within microfluidic channels have been proposed,84–86 though they may impose limitations on the total number of droplets that can be accommodated. Finally, advanced on-chip systems incorporating valves (Fig. 5) have been proposed, enabling the storage and manipulation of millions of droplets.87,88 The latter approach offers a high potential for robustness, automation, and the integration of multiple droplet manipulations onto a single microfluidic chip.

FIG. 5.

Prolonged droplet incubation can be established by implementing on-chip valves. By not fully closing the valve, oil can flow through while droplets are maintained in the collection chamber. The valves can be opened and closed on demand to enable automated workflows. Adapted with permission from Zhou et al., Talanta 253, 124044 (2023). Copyright 2023 Elsevier.

FIG. 5.

Prolonged droplet incubation can be established by implementing on-chip valves. By not fully closing the valve, oil can flow through while droplets are maintained in the collection chamber. The valves can be opened and closed on demand to enable automated workflows. Adapted with permission from Zhou et al., Talanta 253, 124044 (2023). Copyright 2023 Elsevier.

Close modal

Droplet sorting is a frequently used manipulation for the enrichment of droplet populations and is based on the sorting of droplets toward different outlets using passive or active methods. Passive methods rely on (1) hydrodynamic forces to sort droplets by size89–92 or their viscoelastic properties93 or (2) interfacial interactions to sort by interfacial tension.94,95 While these methods offer automation and robustness, they are limited to applications where the physical properties of droplets are directly related to the assay and are, therefore, not broadly applicable.

Active methods, using mainly pneumatic,96 acoustic,97 and dielectrophoretic4,98–100 (DEP)-based actuation, typically offer more versatility due to the potential for integration with a wide range of readout methods allowing for on-demand control. Here, we will focus on the more widely used DEP-based sorters as this actuation method allows highly robust sorting at high accuracies (typically, >99%) and high throughputs, while being compatible with a large range of droplet sizes.101,102 Many research efforts have led to the advancement in sorting throughput by hardware developments and design improvements without compromising accuracy. The major limitation to ultrahigh sorting rates remains the droplet size, as sorting rates of 24,103,104 up to 30 kHz98 have been demonstrated for droplets up to 25 μm in diameter, while droplet sorting of larger droplets of 267 and 337 μm in diameter was limited to 21 and 4 Hz, respectively.105 The accessibility of these tools for non-experts has been improving thanks to the publication of several detailed protocols,106–108 guiding non-trained users through building and operating the droplet sorting platforms. However, current methods mostly rely on manual setting of the operational parameters, which can lead to false sorting in long experiments due to flow perturbations. Therefore, in practice, the user needs to continuously monitor the experiment and adjust flow parameters where needed. In an effort to automate and enhance the robustness of droplet sorting, iSort109 (Fig. 6) was recently introduced, which is based on a closed feedback system that measures the impedance in the channel before and after the sorting junction to obtain information (i.e., spacing, droplet width, and trajectory) on the droplets passing by. With this information, the sorting efficiency and throughput are optimized in real-time by adjusting different parameters affecting the electrical actuation and fluid flow, ultimately lowering the false positives by almost 17-fold.

FIG. 6.

Highly robust droplet sorting by relying on feedback measurements in iSort. Electrical impedance is measured in the main droplet channel and in the collection channel to analyze droplet spacing, width, and trajectory, based on which operational parameters are adjusted in real-time.109 P = pressure, Q = flow rate, fluo = fluorescence, Electr = electrode, Imped = impedance. Adapted with permission from Panwar et al., Cell Rep. Methods 3(5), 100478 (2023). Copyright 2023 Elsevier.

FIG. 6.

Highly robust droplet sorting by relying on feedback measurements in iSort. Electrical impedance is measured in the main droplet channel and in the collection channel to analyze droplet spacing, width, and trajectory, based on which operational parameters are adjusted in real-time.109 P = pressure, Q = flow rate, fluo = fluorescence, Electr = electrode, Imped = impedance. Adapted with permission from Panwar et al., Cell Rep. Methods 3(5), 100478 (2023). Copyright 2023 Elsevier.

Close modal

Advances are also being made in the context of DEP-based multiplex droplet sorting with up to six outlets on one chip.110–112 This has been achieved in two main ways. One approach arranges multiple sorting points in series, allowing sorting up to 3 (Ref. 110) and 700 Hz.113 However, this setup is susceptible to flow disturbances, leading to droplets arriving at the sorting points at different times, which reduces sorting accuracy. The other approach places sorters in parallel, allowing sorting up to 10 (Ref. 114), 100 (Ref. 115), and 200 Hz111 but requires strong electric forces to move the droplets which sometimes lead to droplet splitting. To overcome these challenges, researchers have developed a specialized electrode setup that applies smaller electric forces in steps, thereby creating a larger displacement without the risk of droplet splitting. As such, the multiplex sorting throughput was increased up to 473 Hz in a fiveway configuration.112,116,117 Additionally, a multiplex sorter based on the combination of parallel and serial sorters, allowing sorting up to 80 Hz, was introduced, thereby addressing the major limitations of the individual approaches.118 Ultimately, the combination of the iSort feedback system with multiplex sorters would provide a great opportunity to succeed in automated and robust multiplex droplet sorting.

For many applications, downstream processing of droplets is crucial. Although droplets are commonly recovered after bulk dispensing in a tube,119 certain applications, including specific single-cell studies, require droplet collection at the single droplet level.119,120 The developments in this field are rather recent and require precise coordination between (1) the droplet release, which is mostly based on optical detection,87,121–123 and (2) the substrate position, which is generally enabled by programming a mechanical stage.121,124 In this context, several stand-alone droplet dispensers have been developed that allow to eject droplets into well plates using electrohydrodynamic actuation at rates of about 1 Hz.122,123 However, these dispensers require manual intervention to transfer bulk-collected droplets to the dispenser. Several approaches with increased integration have recently implemented single-droplet dispensing in-line with a DEP-based droplet sorter, by directly printing the droplets onto a substrate with carrier oil at 4 Hz124 or by using pressure-based ejection of the droplets into a tube up to 20 Hz.121 In this context, a recently developed workflow enables droplet sorting, collection of the sorted droplets in a collection chamber on-chip, followed by single-droplet imaging and dispensing in 96-well plates using pressure-based ejection, at a rate of 1 Hz with over 99% accuracy (Fig. 7).87 Next to ensuring robustness in an integrated workflow, this approach provides users with visual evidence on individually dispensed droplets and as such facilitates meaningful post-processing of relevant droplets only (e.g., containing a single cell).

FIG. 7.

Single-droplet dispensing by pressure ejection. In the Cyto-Mine systems of Sphere Fluidics, droplets are monitored in brightfield using a camera to determine droplet content (e.g., single cell or multiple cells) prior to dispensing. A microtiter plate is repositioned for every droplet to enable single-droplet dispensing. Q = flow rate, bf = bright field. Adapted with permission from Josephides et al., SLAS Technol. 25(2), 177–189 (2020). Copyright 2020 Elsevier.

FIG. 7.

Single-droplet dispensing by pressure ejection. In the Cyto-Mine systems of Sphere Fluidics, droplets are monitored in brightfield using a camera to determine droplet content (e.g., single cell or multiple cells) prior to dispensing. A microtiter plate is repositioned for every droplet to enable single-droplet dispensing. Q = flow rate, bf = bright field. Adapted with permission from Josephides et al., SLAS Technol. 25(2), 177–189 (2020). Copyright 2020 Elsevier.

Close modal

For many applications, further exemplified in Sec. V, different droplet manipulations can be combined into functional workflows tailored to specific applications. For non-expert users, integrating these manipulations into a single chip is highly desirable and several approaches to achieve this have been reported.2,83,87,88,125 Aside from reducing user intervention, another clear advantage of chip integration is minimizing droplet coalescence and losses, which often occur during off-chip incubation and reinjection steps. However, several challenges arise when attempting to integrate these workflows into a single chip.

First, delay lines are suitable only for relatively short incubation times (up to 1 h22), while incubation chambers sealed with valves require larger chip footprints when working with extensive droplet libraries. Second, when using delay lines, it is critical to match the droplet speeds between consecutive manipulations, which is challenging because, for example, droplet sorting rates are often slower than droplet generation rates. One possible solution is to decouple these droplet manipulations by incorporating incubation chambers and on-chip valves87,88 between each step. Finally, more complex workflows inherently pose a higher risk of errors. Therefore, real-time monitoring, using techniques like camera-38,87 or impedance-based109 measurements, might be necessary to ensure the robustness of these workflows. Despite these challenges, more integration is certainly achievable, and it will be exciting to observe how future innovations in microfluidic technology push these boundaries further.

In recent years, more droplet microfluidic platforms are becoming commercially available to the end users. First of all, microfluidic chips of various specified droplet size ranges can nowadays be purchased through companies such as Microfluidic ChipShop,126 Micronit,127 Atrandi Biosciences,128 and Dolomite microfluidics,129 either by selecting from their product portfolio or by requesting custom designs. These chips can then be used with commercial syringe pumps (e.g., from CETONI130 and Harvard Apparatus131) or pressure-driven pumps (e.g., from Elveflow,132 Cellix Limited,133 and Fluigent134). Pressure-driven pumps reach equilibrium faster and are able to deliver a constant, non-pulsating flow,37 while syringe pumps allow flow rate programming, which simplifies the microfluidic workflow as flow rates are typically held constant between runs of the same experiment. Pressure-driven pumps can often be integrated with a downstream flow sensor that feeds back to the pressure module, which is of high interest for accurate and responsive flow control. In addition to the pumps, setups for monitoring and readout are required (e.g., a microscope with a high-speed camera and photomultiplier tubes for fluorescent droplet detection).

Therefore, companies are providing more integrated solutions for droplet generation and/or manipulation, as detailed in Table I. For these technologies, there is typically a clear trade-off between flexibility vs robustness and user-friendliness.

TABLE I.

An overview of companies that provide integrated solutions for w/o droplet manipulations. The different enabled droplet manipulations and main features of the platforms are highlighted. Mentioned droplet size ranges and droplet manipulation speeds are indicative as found in online resources. It has to be noted that for the open platforms (all except Chromium, Tapestri, QX600 droplet digital PCR System, Cyto-Mine®, and Cyto-Mine® Chroma), different chips could theoretically be used to achieve droplet sizes outside of the reported range. Additionally, in general, droplet manipulation speeds are highly dependent on the droplet size. Therefore, the reported droplet throughputs cannot necessarily be achieved for all droplet sizes and are not necessarily the highest throughputs that can be achieved with the system.

CompanyPlatformDroplet manipulationsMain features
10X Genomics Chromium >10 000 nanoliter-sized droplets with barcoded beads are generated within 4 min.135 Exact droplet sizes not specified, likely around 90 μm diameter.136  Fully integrated solution for single-cell sequencing of transcriptome, epigenome, intracellular, and cell surface proteins.137  
Mission Bio Tapestri >10 000 nanoliter-sized droplets are generated within 5 min. Afterward, droplets are merged with barcodes within 45 min.138  Fully integrated solution for single-cell sequencing of genome and cell surface proteins.139  
Bio-Rad Laboratories QX600 Droplet Digital PCR System Creates ∼20 000 nanoliter-sized droplets in 2 min.140  Fully integrated solution for digital PCR.141  
Dolomite Microfluidics Micro Droplet Systems (e.g., μEncapsulator) Droplet generation up to 10 kHz in a diameter range of 20–250 μm.142,143 System comprising of high-speed camera, microscope, flow control hardware, and custom microfluidic chips.142  
PreciGenome iFlowTM Droplet generation at multiple kHz with a diameter between 30 and 420 μm.144  System comprising of high-speed camera, microscope, flow control hardware, and custom microfluidic chips.145  
Fluigent and Secoya Complex Emulsion Platform and RayDrop Droplet generation up to 10 kHz in a diameter range of 20–450 μm.146,147 System comprising of high-speed camera and flow control hardware provided by Fluigent, coupled to RayDrop re-usable microfluidic chip from Secoya designed for robust long-term experiments.146,147 
LiveDrop OneFlow Droplet generation up to 14 kHz in a diameter range of 15–over 65 μm.148  Integrated platform with high-speed camera, microscope, and flow control hardware with unique sample loading system that eliminates dead volume.148  
Atrandi Biosciences Onyx Droplet generation in a diameter range of 25–180 μm. Picoinjection, merging, and splitting of similar sized droplets on separate chips.149 Throughput is not mentioned. Integrated platform with high-speed camera, flow control, and electrode control hardware.150  
Atrandi Biosciences Styx Sorting of droplets with a diameter between 30 and 100 μm.149 Throughput of >1 kHz was reported.151  Integrated platform with up to four lasers and four detection colors for fluorescence activated droplet detection and sorting.152  
LiveDrop ModaFlow Droplet generation up to 14 kHz in a diameter range of 15 to over 65 μm. Sorting of the same droplets with a throughput of >1 kHz.153  Integrated platform with up to four lasers and five detection colors for fluorescence activated droplet detection and sorting, as well as a unique sample loading system that eliminates dead volume.153  
Sphere Fluidics Cyto-Mine® and Cyto-Mine® Chroma Generation of droplets with a diameter of about 95 μm at 1 kHz, droplet incubation (0.5–24 h), sorting (200 Hz), and single droplet dispensing (1 Hz) (single integrated chip).87,154 Fully integrated, end-to-end automated single cell analysis platform that can encapsulate, analyze, sort, and dispense viable single cells. The Cyto-Mine® system has one laser and two detection colors, while the Cyto-Mine® Chroma system has four lasers and four detection colors for fluorescence activated droplet sorting.155,156 
CompanyPlatformDroplet manipulationsMain features
10X Genomics Chromium >10 000 nanoliter-sized droplets with barcoded beads are generated within 4 min.135 Exact droplet sizes not specified, likely around 90 μm diameter.136  Fully integrated solution for single-cell sequencing of transcriptome, epigenome, intracellular, and cell surface proteins.137  
Mission Bio Tapestri >10 000 nanoliter-sized droplets are generated within 5 min. Afterward, droplets are merged with barcodes within 45 min.138  Fully integrated solution for single-cell sequencing of genome and cell surface proteins.139  
Bio-Rad Laboratories QX600 Droplet Digital PCR System Creates ∼20 000 nanoliter-sized droplets in 2 min.140  Fully integrated solution for digital PCR.141  
Dolomite Microfluidics Micro Droplet Systems (e.g., μEncapsulator) Droplet generation up to 10 kHz in a diameter range of 20–250 μm.142,143 System comprising of high-speed camera, microscope, flow control hardware, and custom microfluidic chips.142  
PreciGenome iFlowTM Droplet generation at multiple kHz with a diameter between 30 and 420 μm.144  System comprising of high-speed camera, microscope, flow control hardware, and custom microfluidic chips.145  
Fluigent and Secoya Complex Emulsion Platform and RayDrop Droplet generation up to 10 kHz in a diameter range of 20–450 μm.146,147 System comprising of high-speed camera and flow control hardware provided by Fluigent, coupled to RayDrop re-usable microfluidic chip from Secoya designed for robust long-term experiments.146,147 
LiveDrop OneFlow Droplet generation up to 14 kHz in a diameter range of 15–over 65 μm.148  Integrated platform with high-speed camera, microscope, and flow control hardware with unique sample loading system that eliminates dead volume.148  
Atrandi Biosciences Onyx Droplet generation in a diameter range of 25–180 μm. Picoinjection, merging, and splitting of similar sized droplets on separate chips.149 Throughput is not mentioned. Integrated platform with high-speed camera, flow control, and electrode control hardware.150  
Atrandi Biosciences Styx Sorting of droplets with a diameter between 30 and 100 μm.149 Throughput of >1 kHz was reported.151  Integrated platform with up to four lasers and four detection colors for fluorescence activated droplet detection and sorting.152  
LiveDrop ModaFlow Droplet generation up to 14 kHz in a diameter range of 15 to over 65 μm. Sorting of the same droplets with a throughput of >1 kHz.153  Integrated platform with up to four lasers and five detection colors for fluorescence activated droplet detection and sorting, as well as a unique sample loading system that eliminates dead volume.153  
Sphere Fluidics Cyto-Mine® and Cyto-Mine® Chroma Generation of droplets with a diameter of about 95 μm at 1 kHz, droplet incubation (0.5–24 h), sorting (200 Hz), and single droplet dispensing (1 Hz) (single integrated chip).87,154 Fully integrated, end-to-end automated single cell analysis platform that can encapsulate, analyze, sort, and dispense viable single cells. The Cyto-Mine® system has one laser and two detection colors, while the Cyto-Mine® Chroma system has four lasers and four detection colors for fluorescence activated droplet sorting.155,156 

For example, 10X Genomics137 and Mission Bio139 offer highly integrated and robust platforms, yet these are only capable of generating droplets of a fixed size and are highly tailored for application in single-cell omics studies, while Bio-Rad Laboratories141 offers a similarly integrated and robust platform specifically designed for droplet digital PCR.

In contrast, companies such as Dolomite Microfluidics,142 Fluigent,146 Secoya,147 LiveDrop,148 and PreciGenome145 are offering microfluidics application packs for microfluidic droplet generation, consisting of a set of pressure pumps, droplet generation chips, and a high-speed camera. These setups provide increased flexibility since they can be utilized for applications as defined by the user, yet a higher user-interference is required and the platforms are still limited to droplet generation only, which is too limited for many applications.

To provide even more flexibility, devices such as the Onyx150 and Styx152 from Atrandi Biosciences and Modaflow153 from LiveDrop have been developed. These devices allow multiple droplet manipulations and are highly tailorable to the application of interest. Although these platforms are aimed to provide user-friendly workflows, user-interference is still high since manual manipulations are required between different droplet manipulation steps. Therefore, for workflows with multiple subsequent droplet manipulations, skilled personnel are still required.

In this regard, the Cyto-Mine platforms155,156 of Sphere Fluidics are much more integrated since they enable droplet generation, incubation, sorting, and dispensing on a single chip, without requiring any user interference. Additionally, these are currently the only platforms providing single-droplet dispensing. As a downside of this integrated workflow, it is only compatible with droplets of a specific size, and it is not possible to include any other droplet manipulations, such as reagent addition, that might be required for an application.

In conclusion, for commercially available devices, high automation and integration often limit flexibility, making them suitable for specific rather than widespread biological applications. On the other hand, highly integrated platforms enhance the robustness of droplet manipulations, while more flexible systems need constant monitoring and expertise, hindering their use in non-specialized labs. Given the wide variety of options available today, suited systems can be selected depending on the skills and application needs of the users. When searching for the ideal system for a specific application, it is important to look beyond performance metrics such as droplet generation frequency, sorting speed, and sorting accuracy. These metrics can vary significantly depending on factors such as droplet size, the type of bioassay being implemented, and, in some cases, the user's level of expertise. Therefore, it is essential to consider the broader context, including the adaptability of the system to the required workflow and the ease of operation. Whenever possible, arranging a demonstration or trial run of the system can provide valuable insights into its practical performance and help ensure it meets the requirements.

In addition to improving the robustness and user-friendliness of w/o droplet manipulations, ongoing research aims to provide alternatives that enable replacing some or all of the complex manipulations while maintaining several advantages of microfluidic w/o droplets (Fig. 8).

FIG. 8.

Water-in-oil droplets and alternatives.

FIG. 8.

Water-in-oil droplets and alternatives.

Close modal

Some of these are water-in-oil-in-water (w/o/w) droplets and so-called double emulsions. These have one or more aqueous cores and a thin oil shell and are dispersed in an aqueous continuous phase. Generation of double emulsions conventionally is achieved by either two separate microfluidic devices157 or one device with two consecutive droplet generation zones,61,158 having, respectively, hydrophobic and hydrophilic properties for consecutive generation of the w/o and w/o/w emulsions. To overcome the need for the complex localized adjustment of surface wettability in the chip, and thereby render the w/o/w generation more robust, an alternative one-step emulsification procedure was developed, relying on a dual bore capillary system.159 This was later on extended to multiple-bore systems to enable the robust generation of monodisperse double emulsions with up to four inner drops.160 Thanks to the aqueous outer phase of double emulsions, these can be sorted and dispensed with standard, commercially available FACS161 or other dispensers,162 omitting the need for more complex on-chip droplet sorting. The compatibility of double emulsions with FACS implies certain levels of robustness, automation, and accessibility as FACS is commonly used in research environments.157,161 This translates into several published protocols for non-expert users.163,164 Nevertheless, it should be noted that commonly reported post-sort recoveries are low (40–70%),161,165 with a maximal recovery of 81.5% being recently reported166 only after (1) a specialized drop delay calibration procedure, which requires the preparation of dedicated double emulsions comprising the standard AccuDrop beads used for drop delay calibration, and (2) using double emulsions with a limited outer diameter (30 μm) to ensure compatibility with the FACS nozzle size, which limits their use for large mammalian cells or combinations of multiple cells, which need larger droplets for maintaining their cellular activities. In addition, other manipulations such as reagent addition, splitting, and washing are more complicated for double emulsions due to the additional liquid layer, and currently only a few proof-of-concepts of reagent addition167–170 have been described. Nevertheless, double emulsions have been used in a variety of applications, including sorting of sequence-specific DNA molecules through double emulsion digital droplet PCR,165 cellular heterogeneity studies and rare cell sorting,163 and directed evolution of enzymes.157 

Other heavily researched alternatives are hydrogel-based particles, which can be generated by relying on the same principles as w/o droplets: first, droplets of a water-soluble hydrogel precursor are generated using standard droplet generation systems, followed by cross-linking the hydrogel by, e.g., illumination with light and subsequent removal of the oil phase, resulting in hydrogel-based particles resuspended in an aqueous phase. As such, three types of hydrogel-based particles can be generated (1) hydrogel beads, consisting completely of a hydrogel mesh,171 (2) hollow hydrogel capsules (also known as semi-permeable capsules),172 which are liquid cores surrounded by a hydrogel shell, and (3) crescent-shaped hydrogel nanoparticles with a cavity.173–175 Similar to double emulsions, all hydrogel-based particles can be sorted and dispensed with FACS176–179 or similar dispensers,162 although being hampered by the same recovery limitations mentioned above for double emulsions, linked to specialized calibration requirements and limited particle size. In addition, all hydrogel particle types have specific features, rendering them more or less suited for specific applications. For example, for cell studies, it should be noted that cells in hydrogel beads are in close contact with the hydrogel matrix, which is known to impact, e.g., cellular growth and other processes,180 and which is not the case in hydrogel capsules or crescent particles. With respect to the ease of particle generation, hollow hydrogel capsules, and crescent hydrogel particles require precise tuning of the hydrogel precursor compositions, flow rates, and channel geometries to generate stable particles, as they rely on aqueous two phase separation.181 However, the crescent-shaped hydrogel particles can be fabricated in the absence of the sample as they enable subsequent sample encapsulation by mixing using standard lab equipment.175 As such, the microfluidic workflow, required for particle fabrication, is completely decoupled from the biological implementation and can be performed months to years in advance,175 hence obviating the need for any microfluidics expertise for the end user. In terms of reagent exchange and implementation in multi-step assays, all particle types allow the exchange of reagents through the hydrogel mesh or particle cavity by resuspending in alternating aqueous solutions through simple pipetting,172,179 as such facilitating manipulations for the end user in a robust and user-friendly manner. However, with respect to molecule retention, hydrogel beads do not allow for selective retention of components because of the uniform mesh permeability and the lack of a hollow core.172 The retention potential of hollow hydrogel capsules, in turn, can be tuned by changing the shell permeability, but this requires precise control over the composition to not hamper capsule concentricity and stability.172 In addition, while retaining small molecules (e.g., enzymes and short DNA fragments) in capsules remains challenging, this would also result in an overall reduced reagent exchange through the shell, thereby limiting the applicability of these capsules in a range of molecular assays. Finally, retention of cells and molecules of interest in crescent hydrogel particles can only be ensured by capturing them on the particle surface or/and by particle-templated droplet generation.181 Whereas the latter ensures molecule confinement and avoids crosstalk between particles, it also introduces the need for additional de-emulsification steps to subsequently render the particles compatible with standard laboratory methods.172,179

Based on their specifications and limitations, the different particles have been used for a wide range of applications. Hydrogel beads have, for example, been used for rare cell detection182 and as 3D culture platforms,183 and are also at the core of the barcoding methods for single-cell sequencing in Drop-seq,184 InDrop,185 and Hydrop.186 Alternatively, semi-permeable capsules have been demonstrated to enable complex molecular workflows including digital gene expression profiling of individual mammalian cells179 and single genome amplification as well as clonal expansion of bacteria.172 The crescent-shaped particles in turn are commonly used for fluorescence immunoassays on proteins, secreted by individual cells.175,181 However, considering the unique advantages of application of these hydrogel-based particles by non-microfluidic-experts, in combination with future technological advancements that will drastically expand the functionality of these particles in the coming years, they are expected to continue to appear in exciting applications.

Also, for these alternative technologies, commercial platforms are arising. For example, double emulsion and hydrogel bead formulation are supported by the InstaDrop platform of Secoya,147 the Complex emulsion platform of Fluigent146 and the μEncapsulator system by Dolomite Microfluidics.142 Double emulsions are also supported by the Xdrop® Sort system of Samplix.187 Semi-permeable capsules can be generated using the Onyx150 and Flux188 devices from Atrandi Biosciences. Finally, pre-made crescent nanoparticles can be purchased from Partillion Bioscience.189 

Many applications require multistep workflows, where several of the described droplet manipulations must be performed consecutively. In this section, we present three examples of applications that significantly benefit from droplet microfluidics or their alternatives, along with their associated workflows. As such, we aim to inspire researchers with potential workflow ideas for their applications.

The first key application is high-throughput enzyme engineering and discovery, which is extensively reviewed elsewhere.25 Here, single variants from a large protein library (105–108 variants) are encapsulated to identify the best-performing ones, for which two approaches are commonly used. The first approach relies on protein-expressing cells. Here, individual cells expressing protein variants are encapsulated with assay reagents (e.g., fluorogenic substrate for an enzyme and/or lysis buffer) to identify and sort optimal variants. This workflow is most often performed using w/o droplet microfluidics, where droplets are either incubated and sorted on the same chip83,125 or incubated off-chip for re-injection into a sorting chip,83,105,125,190,191 depending on the reaction time. Since the workflow requires only an encapsulation and sorting step, also double emulsions192 and hydrogels178 in combination with FACS are suitable for this application. The second approach relies on gene variants that need to be transcribed and translated into proteins after encapsulation. Here, single gene variants are encapsulated in droplets together with amplification reagents. Multiple subsequent steps have to be performed, including the addition of in vitro transcription and translation reagents, the addition of assay reagents, and sorting.6,193,194 Because many steps are required, current work relies fully on w/o droplet microfluidic workflows, in which the steps are typically performed on separate chips to accommodate lengthy incubation times. In both approaches, the sorted variants are either re-screened for further optimization or sequenced for in vitro production. As such, droplet microfluidics and its alternatives greatly accelerate the discovery of highly efficient enzymes, given the increased throughput compared to classical plate-based assays.

A second application is the field of single-cell omics, where droplet microfluidics is a key technology for single-cell RNA and DNA sequencing, achieving throughputs of thousands of cells.195–198 Typically, single cells are encapsulated with barcoded beads and lysis and/or reverse transcription reagents, resulting in capturing of the cell-specific RNA and/or DNA for downstream analysis. On the microfluidics level, the most commonly used platforms are relatively straightforward since they rely solely on droplet generation. This has resulted in several open source1,186 and commercially available137,139 platforms that are highly specified for these applications. Recent research is delving into using multi-step workflows for increased functionality199 or increased efficiency and sensitivity, for example, by (1) uncoupling different reaction steps using picoinjection200 or (2) processing only cell-containing droplets using either droplet sorting200 or selective merging of those droplets with the barcoded beads.62,87,88,201

A third application is the field of monoclonal antibody discovery, where cells expressing antibodies are screened to identify the antibody-expressing cell of interest for single-cell sequencing and in vitro antibody production. In droplet microfluidic workflows, the cells are co-encapsulated in droplets together with bioassay reagents. After incubation, either on-chip2,87,88 or off-chip,3,88,201,202 the droplets are sorted. Different approaches have been applied to enable the subsequent analysis, for example, by single-droplet dispensing into a microtiter plate for sequencing87,203 or reinjection of all positive cells into a single-cell sequencing chip.201 After sequencing, the antibodies can be produced in vitro for functional testing. As such, large libraries of cells can be screened and rare cells of interest can be isolated, accelerating monoclonal antibody discovery. In this context, the commercially available Cyto-Mine® devices87,155,156 are specifically designed for single-cell phenotype screening applications, such as monoclonal antibody discovery, with as key advantage their capability for single-droplet dispensing, enabling immediate single-cell sequencing. Alternatively, these workflows can also be developed on more open systems like the Onyx150 and Styx,152 or LiveDrop Modaflow153 platforms, albeit with pooled collection of cells of interest. Finally, alternatives to droplet microfluidics have also proven effective for monoclonal antibody discovery, including the encapsulation of assay reagents and antibody-producing cells in full hydrogel beads204 or crescent-shaped hydrogel particles,175 followed by single-particle sorting with FACS, thereby offering a more accessible approach.

We refer the reader to several in-depth reviews9,12,35,205–209 for more examples of possible high-impact applications of droplet microfluidics, such as in the fields of particle synthesis, organoids, synthetic biology, single-cell analysis, personalized medicine, and digital polymerase chain reaction (PCR).

In recent years, the field of droplet microfluidics is increasingly focusing on the implementation of groundbreaking biological or chemical assays inside w/o droplets. Therefore, to improve the reliability and widespread use of these techniques, significant efforts are being made to enhance the robustness of droplet microfluidics on different aspects such as minimizing user-intervention, droplet loss, spontaneous droplet coalescence, and flow instabilities. A clear trend in this context is the implementation of feedback systems, as is seen for, e.g., (1) monodisperse droplet generation by using camera-feedback and real-time adjustments of fluid flow, (2) accurate picoinjection by using oil pressure-feedback and real-time adjustments of the injector Laplace pressure, and (3) robust droplet sorting by impedance-feedback and real-time adjustments of the electrical actuation and fluid flow. Another trend is the integration of multiple droplet manipulations in a single chip to reduce the need for constant user intervention, thereby simplifying experimental workflows and reducing droplet loss. This is mostly obtained by implementing incubation steps in between different manipulations, using either passive delay lines or incubation chambers which can be actively opened or closed. A last aspect is the reliance on passive systems, which can be obtained by clever channel design and has shown suitable for, e.g., delay lines, droplet splitting, and droplet pairing prior to droplet merging. Since the droplet size is highly crucial for passive manipulations, a feedback system on the droplet size might need to be incorporated to ensure a robust workflow. In the future, we anticipate increased integration of microfluidic manipulations, relying on a combination of feedback mechanisms and passive systems.

The trends discussed above are not only observed and foreseen for droplet workflows developed during academic research but also for commercial devices. Namely, boosted by the great interest from a wide range of fields to use droplets, commercialization of w/o droplet generation and other manipulations are increasing, thereby leading to broader adoption of the technique by different end users. The commercial landscape is very broad, ranging from plug-and-play flexible systems that require a lot of user-interference, to highly integrated and robust devices that are tailored to a certain droplet manipulation sequence or even one specific application. This trade-off between robustness and flexibility has to be taken into account by the end user. Namely, when the application aligns with a commercial, integrated workflow, it is often the preferable option. However, when there is no such fit, end users are directed to more open platforms to sequentially implement different droplet manipulations. One of the major challenges in implementing such workflows is the precise design of microfluidic chips, mainly because designs for droplet generation and manipulation are tailored to specific droplet sizes, and certain manipulations, such as reagent addition or splitting, affect the droplet size. In this context, the increasing variety of commercially available chip designs and application packages is expected to enable mix-and-match capabilities for different manipulation units, facilitating a range of functional workflows in the near future. Other efforts to enable faster development of these customizable workflows would be increased sharing of microfluidic designs by the community and the further development of AI-based design tools.210 Moreover, more companies are offering chip fabrication as a service, eliminating the need for microfabrication infrastructure and skills on the part of the end user. On a last note, despite having suitable microfluidic designs, chip operation will still be challenging. Therefore, it is valuable for the community to share common operational principles through protocol papers, which are already becoming more prevalent.

In the context of more straightforward handling by the end user, alternatives such as double emulsions and hydrogel-based particles are arising, enabling non-technical users to benefit from some of the advantages of w/o droplets (e.g., assay miniaturization or single-cell analysis), while using standard laboratory equipment. Given the high interest of end users, we expect further advances in these fields as well, such as increased manipulation possibilities of double emulsions or the use of new hydrogel formulations with different porosities.

As such, we foresee significant advances in both w/o droplets as well as their alternatives that will be mostly focused on the broad adoption of the technologies to further spread their impact throughout different scientific communities.

This work has received funding from KU Leuven (Nos. IDN/20/011, iBOF/23/005, C3/22/046, IOFM/14/002, VTI-24-00161, and C24E/20/035) and Research Foundation Flanders (FWO) (Nos. 12A4W25N, G082522N, S000825N, and G090120N).

The authors have no conflicts to disclose.

Jolien Breukers: Conceptualization (equal); Investigation (equal); Writing – original draft (equal); Writing – review & editing (equal). Karen Ven: Conceptualization (equal); Investigation (equal); Visualization (equal); Writing – original draft (equal); Writing – review & editing (equal). Wannes Verbist: Conceptualization (equal); Investigation (equal); Writing – original draft (equal); Writing – review & editing (equal). Iene Rutten: Conceptualization (equal); Investigation (equal); Writing – original draft (equal); Writing – review & editing (equal). Jeroen Lammertyn: Conceptualization (equal); Supervision (equal); Writing – review & editing (equal).

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

1.
R.
Zilionis
,
J.
Nainys
,
A.
Veres
,
V.
Savova
,
D.
Zemmour
,
A. M.
Klein
, and
L.
Mazutis
, “
Single-cell barcoding and sequencing using droplet microfluidics
,”
Nat. Protoc.
12
(
1
),
44
73
(
2017
).
2.
B.
El Debs
,
R.
Utharala
,
I. V.
Balyasnikova
,
A. D.
Griffiths
,
C. A.
Merten
, and
D. A.
Weitz
, “
Functional single-cell hybridoma screening using droplet-based microfluidics
,”
Proc. Natl. Acad. Sci. U.S.A.
109
(
729
),
11570
11575
(
2012
).
3.
L.
Mazutis
,
J.
Gilbert
,
W. L.
Ung
,
D. A.
Weitz
,
A. D.
Griffiths
, and
J. A.
Heyman
, “
Single-cell analysis and sorting using droplet-based microfluidics
,”
Nat. Protoc.
8
(
5
),
870
891
(
2013
).
4.
D.
Vallejo
,
A.
Nikoomanzar
,
B. M.
Paegel
, and
J. C.
Chaput
, “
Fluorescence-activated droplet sorting for single-cell directed evolution
,”
ACS Synth. Biol.
8
,
1430
1440
(
2019
).
5.
S.
Balasubramanian
,
J.
Chen
,
V.
Wigneswaran
,
C. H.
Bang-Berthelsen
, and
P. R.
Jensen
, “
Droplet-based microfluidic high throughput screening of Corynebacterium glutamicum for efficient heterologous protein production and secretion
,”
Front. Bioeng. Biotechnol.
9
,
668513
(
2021
).
6.
A.
Fallah-Araghi
,
J. C.
Baret
,
M.
Ryckelynck
, and
A. D.
Griffiths
, “
A completely in vitro ultrahigh-throughput droplet-based microfluidic screening system for protein engineering and directed evolution
,”
Lab Chip
12
(
5
),
882
891
(
2012
).
7.
P.
Hammar
,
S. A.
Angermayr
,
S. L.
Sjostrom
,
J.
Van Der Meer
,
K. J.
Hellingwerf
,
E. P.
Hudson
, and
H. N.
Joensson
, “
Single-cell screening of photosynthetic growth and lactate production by cyanobacteria
,”
Biotechnol. Biofuels
8
(
1
),
193
(
2015
).
8.
R.
Gan
,
M. D.
Cabezas
,
M.
Pan
,
H.
Zhang
,
G.
Hu
,
L. G.
Clark
,
M. C.
Jewett
, and
R.
Nicol
, “
High-Throughput regulatory part prototyping and analysis by cell-free protein synthesis and droplet microfluidics
,”
ACS Synth. Biol.
11
(
6
),
2108
2120
(
2022
).
9.
P. C.
Gach
,
K.
Iwai
,
P. W.
Kim
,
N. J.
Hillson
, and
A. K.
Singh
, “
Droplet microfluidics for synthetic biology
,”
Lab Chip
17
,
3388
3400
(
2017
).
10.
A. H. H.
Wong
,
H.
Li
,
Y.
Jia
,
P. I.
Mak
,
R. P. D. S.
Martins
,
Y.
Liu
,
C. M.
Vong
,
H. C.
Wong
,
P. K.
Wong
,
H.
Wang
,
H.
Sun
, and
C. X.
Deng
, “
Drug screening of cancer cell lines and human primary tumors using droplet microfluidics
,”
Sci. Rep.
7
,
9109
(
2017
).
11.
W.
Zhang
,
H.
Jin
,
S.
Lou
,
H.
Yang
,
X.
Dai
, and
S.
Ma
, “
Microfluidic droplet encapsulation-guided organoid growth promotes parental tumor phenotype recapitulation
,”
Int. J. Cancer
154
,
145
154
(
2024
).
12.
S. N.
Lai
,
X.
Ouyang
, and
B.
Zheng
, “
Droplet microfluidics for precision medicine
,” in
Detection Methods in Precision Medicine
, edited by
M.
(Michael) Yang
and
M.
Thompson
(
Royal Society of Chemistry
,
2020
), pp.
253
278
.
13.
A.
Golberg
,
M. L.
Yarmush
, and
T.
Konry
, “
Picoliter droplet microfluidic immunosorbent platform for point-of-care diagnostics of tetanus
,”
Microchim. Acta
180
(
9–10
),
855
860
(
2013
).
14.
K.
Hsieh
,
K. E.
Mach
,
P.
Zhang
,
J. C.
Liao
, and
T. H.
Wang
, “
Combating antimicrobial resistance via single-cell diagnostic technologies powered by droplet microfluidics
,”
Acc. Chem. Res.
55
(
2
),
123
133
(
2022
).
15.
L.
Chen
,
D.
Li
,
X.
Liu
,
Y.
Xie
,
J.
Shan
,
H.
Huang
,
X.
Yu
,
Y.
Chen
,
W.
Zheng
, and
Z.
Li
, “
Point-of-Care blood coagulation assay based on dynamic monitoring of blood viscosity using droplet microfluidics
,”
ACS Sens.
7
(
8
),
2170
2177
(
2022
).
16.
T.
Kawakatsu
,
Y.
Kikuchi
, and
M.
Nakajima
, “
Regular-sized cell creation in microchannel emulsification by visual microprocessing method
,”
J. Am. Oil Chem. Soc.
74
(
3
),
317
321
(
1997
).
17.
Y. C.
Tan
,
J. S.
Fisher
,
A. I.
Lee
,
V.
Cristini
, and
A. P.
Lee
, “
Design of microfluidic channel geometries for the control of droplet volume, chemical concentration, and sorting
,”
Lab Chip
4
(
4
),
292
298
(
2004
).
18.
S.
Takeuchi
,
P.
Garstecki
,
D. B.
Weibel
, and
G. M.
Whitesides
, “
An axisymmetric flow-focusing microfluidic device
,”
Adv. Mater.
17
(
8
),
1067
1072
(
2005
).
19.
J.
Clausell-Tormos
,
D.
Lieber
,
J. C.
Baret
,
A.
El-Harrak
,
O. J.
Miller
,
L.
Frenz
,
J.
Blouwolff
,
K. J.
Humphry
,
S.
Köster
,
H.
Duan
,
C.
Holtze
,
D. A.
Weitz
,
A. D.
Griffiths
, and
C. A.
Merten
, “
Droplet-based microfluidic platforms for the encapsulation and screening of mammalian cells and multicellular organisms
,”
Chem. Biol.
15
(
5
),
427
437
(
2008
).
20.
D. R.
Link
,
S. L.
Anna
,
D. A.
Weitz
, and
H. A.
Stone
, “
Geometrically mediated breakup of drops in microfluidic devices
,”
Phys. Rev. Lett.
92
(
5
),
054503
(
2004
).
21.
K.
Ahn
,
J.
Agresti
,
H.
Chong
,
M.
Marquez
, and
D. A.
Weitz
, “
Electrocoalescence of drops synchronized by size-dependent flow in microfluidic channels
,”
Appl. Phys. Lett.
88
(
26
),
024104
(
2006
).
22.
L.
Frenz
,
K.
Blank
,
E.
Brouzes
, and
A. D.
Griffiths
, “
Reliable microfluidic on-chip incubation of droplets in delay-lines
,”
Lab Chip
9
(
10
),
1344
1348
(
2009
).
23.
J.-C.
Baret
,
O. J.
Miller
,
V.
Taly
,
M.
Ryckelynck
,
A.
El-Harrak
,
L.
Frenz
,
C.
Rick
,
M. L.
Samuels
,
J. B.
Hutchison
,
J. J.
Agresti
,
D. R.
Link
,
D. A.
Weitz
, and
A. D.
Griffiths
, “
Fluorescence-activated droplet sorting (FADS): Efficient microfluidic cell sorting based on enzymatic activity
,”
Lab Chip
9
(
13
),
1817
1972
(
2009
).
24.
K. S.
Elvira
,
F.
Gielen
,
S. S. H.
Tsai
, and
A. M.
Nightingale
, “
Materials and methods for droplet microfluidic device fabrication
,”
Lab Chip
22
(
5
),
859
875
(
2022
).
25.
M.
Gantz
,
S.
Neun
,
E. J.
Medcalf
,
L. D.
van Vliet
, and
F.
Hollfelder
, “
Ultrahigh-throughput enzyme engineering and discovery in in vitro compartments
,”
Chem. Rev.
123
(
9
),
5571
5611
(
2023
).
26.
D. J.
Collins
,
A.
Neild
,
A.
deMello
,
A. Q.
Liu
, and
Y.
Ai
, “
The Poisson distribution and beyond: Methods for microfluidic droplet production and single cell encapsulation
,”
Lab Chip
15
(
17
),
3439
3459
(
2015
).
27.
N.
Shembekar
,
C.
Chaipan
,
R.
Utharala
, and
C. A.
Merten
, “
Droplet-based microfluidics in drug discovery, transcriptomics and high-throughput molecular genetics
,”
Lab Chip
16
(
8
),
1314
1331
(
2016
).
28.
B. E.
Murray
,
L. I.
Penabad
, and
R. T.
Kennedy
, “
Advances in coupling droplet microfluidics to mass spectrometry
,”
Curr. Opin. Biotechnol.
82
,
102962
(
2023
).
29.
Y.
Zhu
and
Q.
Fang
, “
Analytical detection techniques for droplet microfluidics—A review
,”
Anal. Chim. Acta
787
,
24
35
(
2013
).
30.
X.
Xu
,
L.
Cai
,
S.
Liang
,
Q.
Zhang
,
S.
Lin
,
M.
Li
,
Q.
Yang
,
C.
Li
,
Z.
Han
, and
C.
Yang
, “
Digital microfluidics for biological analysis and applications
,”
Lab Chip
23
(
5
),
1169
1191
(
2023
).
31.
L. G.
Welch
,
J.
Estranero
,
P.
Tourlomousis
,
R. C. R.
Wootton
,
V.
Radu
,
C.
González-Fernández
,
T. J.
Puchtler
,
C. M.
Murzeau
,
N. M. G.
Dieckmann
,
A.
Shibahara
,
B. W.
Longbottom
,
C. E.
Bryant
, and
E. L.
Talbot
, “
A programmable and automated optical electrowetting-on-dielectric (oEWOD) driven platform for massively parallel and sequential processing of single cell assay operations
,”
Lab Chip
24
(
16
),
3763
3774
(
2024
).
32.
K.
Samlali
,
F.
Ahmadi
,
A. B. V.
Quach
,
G.
Soffer
, and
S. C. C.
Shih
, “
One cell,oOne drop, one click: Hybrid microfluidics for mammalian single cell isolation
,”
Small
16
(
34
),
2002400
(
2020
).
33.
F.
Ahmadi
,
H.
Tran
,
N.
Letourneau
,
S. R.
Little
,
A.
Fortin
,
A. N.
Moraitis
, and
S. C. C.
Shih
, “
An automated single-cell droplet-digital microfluidic platform for monoclonal antibody discovery
,”
Small
20
(
26
),
2308950
(
2024
).
34.
B. J.
Coelho
,
J. P.
Neto
,
B.
Sieira
,
A. T.
Moura
,
E.
Fortunato
,
R.
Martins
,
P. V.
Baptista
,
R.
Igreja
, and
H.
Águas
, “
Hybrid digital-droplet microfluidic chip for applications in droplet digital nucleic acid amplification: Design, fabrication and characterization
,”
Sensors
23
(
10
),
4927
(
2023
).
35.
T.
Moragues
,
D.
Arguijo
,
T.
Beneyton
,
C.
Modavi
,
K.
Simutis
,
A. R.
Abate
,
J. C.
Baret
,
A. J.
deMello
,
D.
Densmore
, and
A. D.
Griffiths
, “
Droplet-based microfluidics
,”
Nat. Rev. Methods Primers
3
(
1
),
32
(
2023
).
36.
A.
Lashkaripour
,
C.
Rodriguez
,
L.
Ortiz
, and
D.
Densmore
, “
Performance tuning of microfluidic flow-focusing droplet generators
,”
Lab Chip
19
,
1041
1053
(
2019
).
37.
W.
Zeng
,
S.
Li
, and
Z.
Wang
, “
Closed-loop feedback control of droplet formation in a T-junction microdroplet generator
,”
Sens. Actuators, A
233
,
542
547
(
2015
).
38.
D. F.
Crawford
,
C. A.
Smith
, and
G.
Whyte
, “
Image-based closed-loop feedback for highly mono-dispersed microdroplet production
,”
Sci. Rep.
7
(
1
),
10545
(
2017
).
39.
Y.
Xie
,
A. J.
Dixon
,
J. M. R.
Rickel
,
A. L.
Klibanov
, and
J. A.
Hossack
, “
Closed-loop feedback control of microbubble diameter from a flow-focusing microfluidic device
,”
Biomicrofluidics
14
(
3
),
034101
(
2020
).
40.
V.
Bussiere
,
A.
Vigne
,
A.
Link
,
J.
McGrath
,
A.
Srivastav
,
J. C.
Baret
, and
T.
Franke
, “
High-throughput triggered merging of surfactant-stabilized droplet pairs using traveling surface acoustic waves
,”
Anal. Chem.
91
(
21
),
13978
13985
(
2019
).
41.
A. B.
Theberge
,
E.
Mayot
,
A.
El Harrak
,
F.
Kleinschmidt
,
W. T. S.
Huck
, and
A. D.
Griffiths
, “
Microfluidic platform for combinatorial synthesis in picolitre droplets
,”
Lab Chip
12
(
7
),
1320
1326
(
2012
).
42.
L.
Nan
,
T.
Mao
, and
H. C.
Shum
, “
Self-synchronization of reinjected droplets for high-efficiency droplet pairing and merging
,”
Microsyst. Nanoeng.
9
(
1
),
24
(
2023
).
43.
A. R.
Guzman
,
H. S.
Kim
,
P.
de Figueiredo
, and
A.
Han
, “
A three-dimensional electrode for highly efficient electrocoalescence-based droplet merging
,”
Biomed. Microdevices
17
(
2
),
35
(
2015
).
44.
L.
Mazutis
,
J. C.
Baret
, and
A. D.
Griffiths
, “
A fast and efficient microfluidic system for highly selective one-to-one droplet fusion
,”
Lab Chip
9
(
18
),
2665
2672
(
2009
).
45.
L.
Mazutis
and
A. D.
Griffiths
, “
Selective droplet coalescence using microfluidic systems
,”
Lab Chip
12
(
10
),
1800
1806
(
2012
).
46.
X.
Niu
,
S.
Gulati
,
J. B.
Edel
, and
A. J.
Demello
, “
Pillar-induced droplet merging in microfluidic circuits
,”
Lab Chip
8
(
11
),
1837
1841
(
2008
).
47.
H.
Zhang
,
A. R.
Guzman
,
J. A.
Wippold
,
Y.
Li
,
J.
Dai
,
C.
Huang
, and
A.
Han
, “
An ultra high-efficiency droplet microfluidics platform using automatically synchronized droplet pairing and merging
,”
Lab Chip
20
(
21
),
3948
3959
(
2020
).
48.
A. R.
Abate
,
T.
Hung
,
P.
Mary
,
J. J.
Agresti
, and
D. A.
Weitz
, “
High-throughput injection with microfluidics using picoinjectors
,”
Proc. Natl. Acad. Sci. U.S.A.
107
(
45
),
19163
19166
(
2010
).
49.
M.
Rhee
,
Y. K.
Light
,
S.
Yilmaz
,
P. D.
Adams
,
D.
Saxena
,
R. J.
Meagher
, and
A. K.
Singh
, “
Pressure stabilizer for reproducible picoinjection in droplet microfluidic systems
,”
Lab Chip
14
(
23
),
4533
4539
(
2014
).
50.
J.
Breukers
,
H.
Op de Beeck
,
I.
Rutten
,
M.
López Fernández
,
S.
Eyckerman
, and
J.
Lammertyn
, “
Highly flexible and accurate serial picoinjection in droplets by combined pressure and flow rate control
,”
Lab Chip
22
(
18
),
3475
3488
(
2022
).
51.
C.
Frey
,
J. A.
De Lora
,
T.
Jahnke
,
Y.
Wang
,
S.
Weber
,
I.
Platzman
, and
J. P.
Spatz
, “Controlled microfluidic droplet acoustoinjection on one chip,” ChemRxiv (n.d.).
52.
H.
Yuan
,
Y.
Chao
,
S. S.
Li
,
M. Y. H.
Tang
,
Y.
Huang
,
Y.
Che
,
A. S. T.
Wong
,
T.
Zhang
, and
H. C.
Shum
, “
Picoinjection-enabled multitarget loop-mediated isothermal amplification for detection of foodborne pathogens
,”
Anal. Chem.
90
(
22
),
13173
13177
(
2018
).
53.
S. L.
Sjostrom
,
H. N.
Joensson
, and
H. A.
Svahn
, “
Multiplex analysis of enzyme kinetics and inhibition by droplet microfluidics using picoinjectors
,”
Lab Chip
13
(
9
),
1754
1761
(
2013
).
54.
E. X.
Ng
,
M. A.
Miller
,
T.
Jing
,
D. A.
Lauffenburger
, and
C. H.
Chen
, “
Low-volume multiplexed proteolytic activity assay and inhibitor analysis through a pico-injector array
,”
Lab Chip
15
(
4
),
1153
1159
(
2015
).
55.
C. H.
Chen
,
M. A.
Miller
,
A.
Sarkar
,
M. T.
Beste
,
K. B.
Isaacson
,
D. A.
Lauffenburger
,
L. G.
Griffith
, and
J.
Han
, “
Multiplexed protease activity assay for low-volume clinical samples using droplet-based microfluidics and its application to endometriosis
,”
J. Am. Chem. Soc.
135
(
5
),
1645
1648
(
2013
).
56.
S.
Li
,
M.
Zeng
,
T.
Gaule
,
M. J.
McPherson
, and
F. C.
Meldrum
, “
Passive picoinjection enables controlled crystallization in a droplet microfluidic device
,”
Small
13
(
41
),
1702154
(
2017
).
57.
H.
Yuan
,
Y.
Pan
,
J.
Tian
,
Y.
Chao
,
J.
Li
, and
H.
Cheung Shum
, “
Electricity-free picoinjection assisted droplet microfluidics
,”
Sens. Actuators, B
298
,
126766
(
2019
).
58.
X.
Pan
,
S.
Zeng
,
Q.
Zhang
,
B.
Lin
, and
J.
Qin
, “
Sequential microfluidic droplet processing for rapid DNA extraction
,”
Electrophoresis
32
(
23
),
3399
3405
(
2011
).
59.
Z.
Zhao
,
H.
Zhai
,
P.
Zuo
,
T.
Wang
,
R.
Xie
,
M.
Tian
,
R.
Song
,
X.
Xu
, and
Z.
Li
, “
Image-activated pico-injection for single-cell analysis
,”
Talanta
272
,
125765
(
2024
).
60.
A.
Autour
and
C. A.
Merten
, “
Fluorescence-activated droplet sequencing (FAD-seq) directly provides sequences of screening hits in antibody discovery
,”
Proc. Natl. Acad. Sci. U.S.A.
121
(
37
),
e2405342121
(
2024
).
61.
A. R.
Abate
and
D. A.
Weitz
, “
Faster multiple emulsification with drop splitting
,”
Lab Chip
11
(
11
),
1911
1915
(
2011
).
62.
I. C.
Clark
,
C. L.
Delley
,
C.
Sun
,
R.
Thakur
,
S. L.
Stott
,
S.
Thaploo
,
Z.
Li
,
F. J.
Quintana
, and
A. R.
Abate
, “
Targeted single-cell RNA and DNA sequencing with fluorescence-activated droplet merger
,”
Anal. Chem.
92
(
21
),
14616
14623
(
2020
).
63.
D.
Wong
and
C. L.
Ren
, “
Microfluidic droplet trapping, splitting and merging with feedback controls and state space modelling
,”
Lab Chip
16
(
17
),
3317
3329
(
2016
).
64.
S. N.
Agnihotri
,
M. R.
Raveshi
,
R.
Bhardwaj
, and
A.
Neild
, “
Microvalves for integrated selective droplet generation, splitting and merging on a chip
,”
Microfluid. Nanofluid.
25
(
11
),
88
(
2021
).
65.
M. R.
Raveshi
,
S. N.
Agnihotri
,
M.
Sesen
,
R.
Bhardwaj
, and
A.
Neild
, “
Selective droplet splitting using single layer microfluidic valves
,”
Sens. Actuators, B
292
,
233
240
(
2019
).
66.
J. H.
Jung
,
G.
Destgeer
,
B.
Ha
,
J.
Park
, and
H. J.
Sung
, “
On-demand droplet splitting using surface acoustic waves
,”
Lab Chip
16
(
17
),
3235
3243
(
2016
).
67.
P.
Zhang
,
A.
Kaushik
,
K.
Hsieh
, and
T. H.
Wang
, “
Customizing droplet contents and dynamic ranges via integrated programmable picodroplet assembler
,”
Microsyst. Nanoeng.
5
,
22
(
2019
).
68.
T. S.
Kaminski
,
S.
Jakiela
,
M. A.
Czekalska
,
W.
Postek
, and
P.
Garstecki
, “
Automated generation of libraries of nL droplets
,”
Lab Chip
12
(
20
),
3995
4002
(
2012
).
69.
A. C.
Hatch
,
J. S.
Fisher
,
A. R.
Tovar
,
A. T.
Hsieh
,
R.
Lin
,
S. L.
Pentoney
,
D. L.
Yang
, and
A. P.
Lee
, “
1-Million droplet array with wide-field fluorescence imaging for digital PCR
,”
Lab Chip
11
(
22
),
3838
3845
(
2011
).
70.
B.
Verbruggen
,
T.
Tóth
,
Y. T.
Atalay
,
F.
Ceyssens
,
P.
Verboven
,
R.
Puers
,
B.
Nicolai
, and
J.
Lammertyn
, “
Design of a flow controlled asymmetric droplet splitter using computational fluid dynamics
,”
Microfluid. Nanofluid.
15
,
243
252
(
2013
).
71.
W.
Postek
,
T. S.
Kaminski
, and
P.
Garstecki
, “
A passive microfluidic system based on step emulsification allows the generation of libraries of nanoliter-sized droplets from microliter droplets of varying and known concentrations of a sample
,”
Lab Chip
17
(
7
),
1323
1331
(
2017
).
72.
D. R.
Emerson
,
K.
Cieślicki
,
X.
Gu
, and
R. W.
Barber
, “
Biomimetic design of microfluidic manifolds based on a generalised Murray’s law
,”
Lab Chip
6
(
3
),
447
454
(
2006
).
73.
D. A.
Holland-Moritz
,
M. K.
Wismer
,
B. F.
Mann
,
I.
Farasat
,
P.
Devine
,
E. D.
Guetschow
,
I.
Mangion
,
C. J.
Welch
,
J. C.
Moore
,
S.
Sun
, and
R. T.
Kennedy
, “
Mass activated droplet sorting (MADS) enables high-throughput screening of enzymatic reactions at nanoliter scale
,”
Angew. Chem.
132
(
11
),
4500
4507
(
2020
).
74.
X.
Li
and
C.
Smith
, “ESI-MineTM: Picodroplet mass spectrometry for high throughput single cell analysis” (last accessed February 24, 2025), see https://spherefluidics.com/wp-content/uploads/2019/08/ESI-Mine-Picodorplet-Mass-Spectrometry-for-High-Throughput-Single-Cell-Analysis-Advanced-Proteomics-2019-Poster.pdf
75.
W.
Stephenson
, “
High-throughput magnetic particle washing in nanoliter droplets using serial injection and splitting
,”
Micro Nano Syst. Lett.
6
(
1
),
3
(
2018
).
76.
C.
Huang
,
H.
Zhang
,
S. I.
Han
, and
A.
Han
, “
Cell washing and solution exchange in droplet microfluidic systems
,”
Anal. Chem.
93
(
24
),
8622
8630
(
2021
).
77.
B.
Verbruggen
,
K.
Leirs
,
R.
Puers
, and
J.
Lammertyn
, “
Selective DNA extraction with microparticles in segmented flow
,”
Microfluid. Nanofluid.
18
(
2
),
293
303
(
2015
).
78.
Z.
Liu
,
A.
Fornell
,
L.
Barbe
,
K.
Hjort
, and
M.
Tenje
, “
On-chip background dilution in droplets with high particle recovery using acoustophoresis
,”
Biomicrofluidics
13
(
6
),
064123
(
2019
).
79.
J.
Park
,
G.
Destgeer
,
H.
Kim
,
Y.
Cho
, and
H. J.
Sung
, “
In-droplet microparticle washing and enrichment using surface acoustic wave-driven acoustic radiation force
,”
Lab Chip
18
(
19
),
2936
2945
(
2018
).
80.
M. J.
Siedlik
and
D.
Issadore
, “
Pico-washing: Simultaneous liquid addition and removal for continuous-flow washing of microdroplets
,”
Microsyst. Nanoeng.
8
(
1
),
46
(
2022
).
81.
S. R.
Doonan
,
M.
Lin
, and
R. C.
Bailey
, “
Droplet CAR-Wash: Continuous picoliter-scale immunocapture and washing
,”
Lab Chip
19
(
9
),
1589
1598
(
2019
).
82.
H.
Lee
,
L.
Xu
, and
K. W.
Oh
, “
Droplet-based microfluidic washing module for magnetic particle-based assays
,”
Biomicrofluidics
8
(
4
),
044113
(
2014
).
83.
R.
Obexer
,
A.
Godina
,
X.
Garrabou
,
P. R. E.
Mittl
,
D.
Baker
,
A. D.
Griffiths
, and
D.
Hilvert
, “
Emergence of a catalytic tetrad during evolution of a highly active artificial aldolase
,”
Nat. Chem.
9
(
1
),
50
56
(
2017
).
84.
A.
Huebner
,
D.
Bratton
,
G.
Whyte
,
M.
Yang
,
A. J.
Demello
,
C.
Abell
, and
F.
Hollfelder
, “
Static microdroplet arrays: A microfluidic device for droplet trapping, incubation and release for enzymatic and cell-based assays
,”
Lab Chip
9
(
5
),
692
698
(
2009
).
85.
M. A.
Khorshidi
,
P. K. P.
Rajeswari
,
C.
Wählby
,
H. N.
Joensson
, and
H.
Andersson Svahn
, “
Automated analysis of dynamic behavior of single cells in picoliter droplets
,”
Lab Chip
14
(
5
),
931
937
(
2014
).
86.
A.
Kulesa
,
J.
Kehe
,
J. E.
Hurtado
,
P.
Tawde
, and
P. C.
Blainey
, “
Combinatorial drug discovery in nanoliter droplets
,”
Proc. Natl. Acad. Sci. U.S.A.
115
(
26
),
6685
6690
(
2018
).
87.
D.
Josephides
,
S.
Davoli
,
W.
Whitley
,
R.
Ruis
,
R.
Salter
,
S.
Gokkaya
,
M.
Vallet
,
D.
Matthews
,
G.
Benazzi
,
E.
Shvets
,
F.
Gesellchen
,
D.
Geere
,
X.
Liu
,
X.
Li
,
B.
Mackworth
,
W.
Young
,
Z.
Owen
,
C.
Smith
,
D.
Starkie
,
J.
White
,
B.
Sweeney
,
M.
Hinchliffe
,
S.
Tickle
,
D. J.
Lightwood
,
M.
Rehak
,
F. F.
Craig
, and
D.
Holmes
, “
Cyto-Mine: An integrated, picodroplet system for high-throughput single-cell analysis, sorting, dispensing, and monoclonality assurance
,”
SLAS Technol.
25
(
2
),
177
189
(
2020
).
88.
Y.
Zhou
,
Z.
Yu
,
M.
Wu
,
Y.
Lan
,
C.
Jia
, and
J.
Zhao
, “
Single-cell sorting using integrated pneumatic valve droplet microfluidic chip
,”
Talanta
253
,
124044
(
2023
).
89.
H. N.
Joensson
,
M.
Uhlén
, and
H. A.
Svahn
, “
Droplet size based separation by deterministic lateral displacement—Separating droplets by cell-induced shrinking
,”
Lab Chip
11
(
7
),
1305
1310
(
2011
).
90.
D. H.
Yoon
,
D.
Tanaka
,
T.
Sekiguchi
, and
S.
Shoji
, “
Size-dependent and property-independent passive microdroplet sorting by droplet transfer on dot rails
,”
Micromachines
9
,
513
(
2018
).
91.
L.
Mazutis
and
A. D.
Griffiths
, “
Preparation of monodisperse emulsions by hydrodynamic size fractionation
,”
Appl. Phys. Lett.
95
(
20
),
204103
(
2009
).
92.
S.
Numakunai
,
A.
Jamsaid
,
D. H.
Yoon
,
T.
Sekiguchi
, and
S.
Shoji
, “
Multiple size-oriented passive droplet sorting device and basic approach for digital chemical synthesis
,” in
Proceedings of the IEEE International Conference on Micro Electro Mechanical Systems (MEMS)
(IEEE,
2014
), pp.
92
95
.
93.
A. C.
Hatch
,
A.
Patel
,
N. R.
Beer
, and
A. P.
Lee
, “
Passive droplet sorting using viscoelastic flow focusing
,”
Lab Chip
13
(
7
),
1308
1315
(
2013
).
94.
C.
Dobson
,
C.
Zielke
,
C. W.
Pan
,
C.
Feit
, and
P.
Abbyad
, “
Method for passive droplet sorting after photo-tagging
,”
Micromachines
11
(
11
),
964
(
2020
).
95.
C. W.
Pan
,
D. G.
Horvath
,
S.
Braza
,
T.
Moore
,
A.
Lynch
,
C.
Feit
, and
P.
Abbyad
, “
Sorting by interfacial tension (SIFT): Label-free selection of live cells based on single-cell metabolism
,”
Lab Chip
19
(
8
),
1344
1351
(
2019
).
96.
R.
Zhang
,
C.
Gao
,
L.
Tian
,
R.
Wang
,
J.
Hong
,
M.
Gao
, and
L.
Gui
, “
Dynamic pneumatic rails enabled microdroplet manipulation
,”
Lab Chip
21
(
1
),
105
112
(
2021
).
97.
E. S.
Richter
,
A.
Link
,
J. S.
McGrath
,
R. W.
Sparrow
,
M.
Gantz
,
E. J.
Medcalf
,
F.
Hollfelder
, and
T.
Franke
, “
Sorting of droplets at kHz rates using absorbance activated acoustic sorting
,”
Lab Chip
23
,
195
202
(
2022
).
98.
A.
Sciambi
and
A. R.
Abate
, “
Accurate microfluidic sorting of droplets at 30 kHz
,”
Lab Chip
15
(
1
),
47
51
(
2015
).
99.
S. W.
Lim
and
A. R.
Abate
, “
Ultrahigh-throughput sorting of microfluidic drops with flow cytometry
,”
Lab Chip
13
(
23
),
4563
4572
(
2013
).
100.
K.
Ahn
,
C.
Kerbage
,
T. P.
Hunt
,
R. M.
Westervelt
,
D. R.
Link
, and
D. A.
Weitz
, “
Dielectrophoretic manipulation of drops for high-speed microfluidic sorting devices
,”
Appl. Phys. Lett.
88
(
2
),
024104
(
2006
).
101.
H. D.
Xi
,
H.
Zheng
,
W.
Guo
,
A. M.
Gañán-Calvo
,
Y.
Ai
,
C. W.
Tsao
,
J.
Zhou
,
W.
Li
,
Y.
Huang
,
N. T.
Nguyen
, and
S. H.
Tan
, “
Active droplet sorting in microfluidics: A review
,”
Lab Chip
17
(
5
),
751
771
(
2017
).
102.
C.
Huang
,
Y.
Jiang
,
Y.
Li
, and
H.
Zhang
, “
Droplet detection and sorting system in microfluidics: A review
,”
Micromachines
14
(
1
),
103
(
2023
).
103.
J. J.
Agresti
,
E.
Antipov
,
A. R.
Abate
,
K.
Ahn
,
A. C.
Rowat
,
J. C.
Baret
,
M.
Marquez
,
A. M.
Klibanov
,
A. D.
Griffiths
, and
D. A.
Weitz
, “
Ultrahigh-throughput screening in drop-based microfluidics for directed evolution
,”
Proc. Natl. Acad. Sci. U.S.A.
107
(
9
),
4004
4009
(
2010
).
104.
P. Y.
Colin
,
B.
Kintses
,
F.
Gielen
,
C. M.
Miton
,
G.
Fischer
,
M. F.
Mohamed
,
M.
Hyvönen
,
D. P.
Morgavi
,
D. B.
Janssen
, and
F.
Hollfelder
, “
Ultrahigh-throughput discovery of promiscuous enzymes by picodroplet functional metagenomics
,”
Nat. Commun.
6
(
1
),
10008
(
2015
).
105.
T.
Beneyton
,
I. P. M.
Wijaya
,
P.
Postros
,
M.
Najah
,
P.
Leblond
,
A.
Couvent
,
E.
Mayot
,
A. D.
Griffiths
, and
A.
Drevelle
, “
High-throughput screening of filamentous fungi using nanoliter-range droplet-based microfluidics
,”
Sci. Rep.
6
,
27223
(
2016
).
106.
F.
Gielen
,
P.-Y.
Colin
,
P.
Mair
, and
F.
Hollfelder
, “
Ultrahigh-throughput screening of single-cell lysates for directed evolution and functional metagenomics
,” in
Methods in Molecular Biology Protein Engineering
, edited by
M. J.
Walker
(
Springer Protocols
,
2014
), pp.
297
310
.
107.
L.
Mazutis
,
J.
Gilbert
,
W. L.
Ung
,
D. A.
Weitz
,
A. D.
Griffiths
, and
J. A.
Heyman
, “
Single-cell analysis and sorting using droplet-based microfluidics
,”
Nat. Protoc.
8
(
5
),
870
(
2013
).
108.
J.
Panwar
,
A.
Autour
, and
C. A.
Merten
, “
Design and construction of a microfluidics workstation for high-throughput multi-wavelength fluorescence and transmittance activated droplet analysis and sorting
,”
Nat. Protoc.
18
(
4
),
1090
1136
(
2023
).
109.
J.
Panwar
,
R.
Utharala
,
L.
Fennelly
,
D.
Frenzel
, and
C. A.
Merten
, “
Isort enables automated complex microfluidic droplet sorting in an effort to democratize technology
,”
Cell Rep. Methods
3
(
5
),
100478
(
2023
).
110.
D.
Frenzel
and
C. A.
Merten
, “
Microfluidic train station: Highly robust and multiplexable sorting of droplets on electric rails
,”
Lab Chip
17
(
6
),
1024
1030
(
2017
).
111.
O.
Caen
,
S.
Schütz
,
M. S. S.
Jammalamadaka
,
J.
Vrignon
,
P.
Nizard
,
T. M.
Schneider
,
J. C.
Baret
, and
V.
Taly
, “
High-throughput multiplexed fluorescence-activated droplet sorting
,”
Microsyst. Nanoeng.
4
(
1
),
33
(
2018
).
112.
A.
Isozaki
,
D.
Huang
,
Y.
Nakagawa
, and
K.
Goda
, “
Dual sequentially addressable dielectrophoretic array for high-throughput, scalable, multiplexed droplet sorting
,”
Microfluid. Nanofluid.
25
(
4
),
32
(
2021
).
113.
S.
Vyawahare
,
M.
Brundage
,
A.
Kijac
,
M.
Gutierrez
,
M.
De Geus
,
S.
Sinha
, and
A.
Homyk
, “
Sorting droplets into many outlets
,”
Lab Chip
21
(
21
),
4262
4273
(
2021
).
114.
M.
Girault
,
H.
Kim
,
H.
Arakawa
,
K.
Matsuura
,
M.
Odaka
,
A.
Hattori
,
H.
Terazono
, and
K.
Yasuda
, “
An on-chip imaging droplet-sorting system: A real-time shape recognition method to screen target cells in droplets with single cell resolution
,”
Sci. Rep.
7
,
40072
(
2017
).
115.
M. E.
Blaha
,
S.
Hasan
,
C.
Dusny
, and
D.
Belder
, “
Fluorescence lifetime activated droplet sorting (FLADS) for label-free sorting of Synechocystis sp. PCC6803
,”
Lab Chip
22
(
8
),
1604
1614
(
2022
).
116.
A.
Isozaki
,
Y.
Nakagawa
,
M. H.
Loo
,
Y.
Shibata
,
N.
Tanaka
,
D. L.
Setyaningrum
,
J. W.
Park
,
Y.
Shirasaki
,
H.
Mikami
,
D.
Huang
,
H.
Tsoi
,
C. T.
Riche
,
T.
Ota
,
H.
Miwa
,
Y.
Kanda
,
T.
Ito
,
K.
Yamada
,
O.
Iwata
,
K.
Suzuki
,
S.
Ohnuki
,
Y.
Ohya
,
Y.
Kato
,
T.
Hasunuma
,
S.
Matsusaka
,
M.
Yamagishi
,
M.
Yazawa
,
S.
Uemura
,
K.
Nagasawa
,
H.
Watarai
,
D.
Di Carlo
, and
K.
Goda
, “
Sequentially addressable dielectrophoretic array for high-throughput sorting of large-volume biological compartments
,”
Sci. Adv.
6
(
22
),
eaba6712
(
2020
).
117.
M. H.
Loo
,
Y.
Nakagawa
,
S. H.
Kim
,
A.
Isozaki
, and
K.
Goda
, “
High-throughput sorting of nanoliter droplets enabled by a sequentially addressable dielectrophoretic array
,”
Electrophoresis
43
(
3
),
477
486
(
2022
).
118.
W.
Verbist
,
J.
Breukers
,
S.
Sharma
,
I.
Rutten
,
H.
Gerstmans
,
L.
Coelmont
,
F.
Dal Dosso
,
K.
Dallmeier
, and
J.
Lammertyn
, “
Separate: Multiway fluorescence-activated droplet sorting based on integration of serial and parallel triaging concepts
,”
Lab Chip
24
,
2107
(
2024
).
119.
P.
Zhang
,
C.
Liu
,
C.
Modavi
,
A.
Abate
, and
H.
Chen
, “
Printhead on a chip: Empowering droplet-based bioprinting with microfluidics
,”
Trends Biotechnol.
42
(
3
),
353
368
(
2024
).
120.
G.
Sun
,
L.
Qu
,
F.
Azi
,
Y.
Liu
,
J.
Li
,
X.
Lv
,
G.
Du
,
J.
Chen
,
C. H.
Chen
, and
L.
Liu
, “
Recent progress in high-throughput droplet screening and sorting for bioanalysis
,”
Biosens. Bioelectron.
225
,
115107
(
2023
).
121.
L.
Nan
,
M. Y. A.
Lai
,
M. Y. H.
Tang
,
Y. K.
Chan
,
L. L. M.
Poon
, and
H. C.
Shum
, “
On-demand droplet collection for capturing single cells
,”
Small
16
(
9
),
1902889
(
2020
).
122.
Y.
Qin
,
L.
Wu
,
J.
Wang
,
R.
Han
,
J.
Shen
,
J.
Wang
,
S.
Xu
,
A. L.
Paguirigan
,
J. L.
Smith
,
J. P.
Radich
, and
D. T.
Chiu
, “
A fluorescence-activated single-droplet dispenser for high accuracy single-droplet and single-cell sorting and dispensing
,”
Anal. Chem.
91
(
10
),
6815
6819
(
2019
).
123.
A.
Ge
,
Z.
Diao
,
Y.
Li
,
C.
Cui
,
X.
Wang
,
Y.
Liu
,
J.
Xu
, and
B.
Ma
, “
An integrated microfluidic platform for on-demand single droplet dispenser with high accuracy by electrohydrodynamic (EHD) printing technique
,”
Sens. Actuators, B
405
,
135334
(
2024
).
124.
R. H.
Cole
,
S. Y.
Tang
,
C. A.
Siltanen
,
P.
Shahi
,
J. Q.
Zhang
,
S.
Poust
,
Z. J.
Gartner
, and
A. R.
Abate
, “
Printed droplet microfluidics for on demand dispensing of picoliter droplets and cells
,”
Proc. Natl. Acad. Sci. U.S.A.
114
(
33
),
8728
8733
(
2017
).
125.
A.
Debon
,
M.
Pott
,
R.
Obexer
,
A. P.
Green
,
L.
Friedrich
,
A. D.
Griffiths
, and
D.
Hilvert
, “
Ultrahigh-throughput screening enables efficient single-round oxidase remodelling
,”
Nat. Catal.
2
(
9
),
740
747
(
2019
).
126.
Microfluidic ChipShop
, see https://www.microfluidic-chipshop.com/catalogue/microfluidic-chips/polymer-chips/ for “Ready-to-Go Microfluidic Chips”(last accessed April 10, 2024).
127.
Micronit
, see https://micronit.com/about-us for “Bring Your Ideas to Life,” (last accessed April 10, 2024).
128.
Atrandi Biosciences
, see https://atrandi.com/microfluidic-chips for “Accelerate Your Microfluidic Research With Reliable Microfluidic Chips” (last accessed April 10, 2024).
129.
Dolomite Microfluidics
, see https://www.dolomite-microfluidics.com/wp-content/uploads/Dolomite-Microfluidic-Chips.pdf for “Mitos Chips” (last accessed April 10, 2024).
130.
CETONI
, see https://cetoni.com/syringe-pump-and-flow-systems/ for “Syringe Pump” (last accessed April 10, 2024).
131.
Harvard Apparatus
, see https://www.harvardapparatus.com/pumps-liquid-handling/syringe-pumps/pump-controller.html for “Pump Controller” (last accessed April 10, 2024).
132.
Elveflow
, see https://www.elveflow.com/microfluidic-products/microfluidics-flow-control-systems/ for “Flow Control Systems” (last accessed April 10, 2024).
133.
Cellix
, see https://www.wearecellix.com/microfluidicpumps for “Microfluidic Pumps” (last accessed April 10, 2024).
134.
Fluigent
, see https://www.elveflow.com/microfluidic-products/microfluidics-flow-control-systems/ for “Microfluidic Instruments,” (last accessed April 10, 2024).
135.
10X Genomics
, see https://www.10xgenomics.com/platforms/chromium/technology for “Chromium GEM-X Technology Single Cell Innovations & Workflows Built for Your Success” (last accessed February 8, 2025).
136.
B.
Wu
,
H. M.
Bennett
,
X.
Ye
,
A.
Sridhar
,
C.
Eidenschenk
,
C.
Everett
,
E. V.
Nazarova
,
H.-H.
Chen
,
I. K.
Kim
,
M.
Deangelis
,
L. A.
Owen
,
C.
Chen
,
J.
Lau
,
M.
Shi
,
J. M.
Lund
,
A.
Xavier-Magalhães
,
N.
Patel
,
Y.
Liang
,
Z.
Modrusan
, and
S.
Darmanis
, “
Overloading and unpacking (OAK)—Droplet-based combinatorial indexing for ultra-high throughput single-cell multiomic profiling
,”
Nat. Commun.
15
,
9146
(
2024
).
137.
10X Genomics
, see https://www.10xgenomics.com/platforms/chromium/product-family for “Chromium Single Cell the Most Versatile Single Cell Toolkit” (last accessed February 8, 2025).
138.
Mission Bio
, see https://missionbio.com/wp-content/uploads/2023/08/Tapestri-Single-Cell-DNA-Protein-Sequencing-v3-User-Guide_MB05-0018.pdf for “Tapestri® Single-Cell DNA + Protein Sequencing v3Tapestri® Single-Cell DNA + Protein Sequencing v3” (last accessed February 8, 2025).
139.
Mission Bio
, see https://missionbio.com/products/platform/ for “Using a Single Cell Sequencing Platform to Untangle the Complexity of Cancer with a True Multi-Omics Approach” (last accessed April 10, 2024).
140.
Bio-Rad
, see https://www.bio-rad.com/webroot/web/pdf/lsr/literature/10031907.pdf for “QX200TM Droplet Generator” (last accessed February 8, 2025).
141.
Bio-Rad
, see https://www.bio-rad.com/en-be/product/qx600-droplet-digital-pcr-system?ID=b07d12ac-0585-fc4c-a586-3ddf20d5c4a0 for “QX600 Droplet Digital PCR System” (last accessed February 8, 2025).
142.
Dolomite Microfluidics
, see https://www.dolomite-microfluidics.com/microfluidic-systems/%c2%b5encapsulator/μEncapsulator Systems” (last accessed April 10, 2024).
143.
Dolomite Microfluidics
, see https://www.dolomite-microfluidics.com/wp-content/uploads/Micro_Droplet_Systems_low_res.pdf for “Mitos Standard Microfluidic Products” (accessed February 8, 2025).
144.
PreciGenome
, see https://www.precigenome.com/_files/ugd/ec0ae9_7825d6fcecc34cbc946709d50ff16898.pdf?index=true “iFlowTM Droplet Generation Application (v1.5)” (last accessed February 8, 2025).
145.
PreciGenome
, see https://www.precigenome.com/microfluidic-fluidic/microfluidic-droplet-generator for “iFlowTM Droplet Generator w. Microfluidic Droplet Generation Chips” (last accessed February 8, 2025).
146.
Fluigent
, see https://www.fluigent.com/research/instruments/packages/complex-emulsion-production-platform/ for “Microfluidic Complex Emulsion Production Platform” (last accessed February 8, 2025).
147.
Secoya
, see https://www.secoya-tech.com/products/raydrop-droplet-generator/ for “Raydrop Droplet Generator” (last accessed February 8, 2025).
148.
LiveDrop
, see https://www.livedrop-bio.com/products/oneflow for “OneFlow” (last accessed February 8, 2025).
149.
Atrandi Biosciences
, see https://atrandi.com/documents/AtrandiBiosciences-Product-Catalogue.pdf for “Product Catalogue” (last accessed February 8, 2025).
150.
Atrandi Biosciences, see https://atrandi.com/products/onyx for “Onyx” (last accessed April 10, 2024).
151.
Atrandi Biosciences
, see https://atrandi.com/documents/DropletGenomics-ApplicationNote-Directed-Evolution.pdf for “Application Note Droplet-based High-Throughput Screening Systems Facilitate In Vitro Evolution of Enzymes” (last accessed February 8, 2025).
152.
Atrandi Biosciences
, see https://atrandi.com/products/styx for “Styx” (last accessed April 10, 2024).
153.
LiveDrop
, see https://www.livedrop-bio.com/products/modaflow for “ModaFlow” (last accessed February 8, 2025).
154.
Sphere Fluidics
, see https://spherefluidics.com/wp-content/uploads/2017/11/Cyto-Mine-Brochure-2017-11-24.pdf “Cyto-Mine® The Single Cell Analysis and Monoclonality Assurance System” (last accessed February 8, 2025).
155.
Sphere Fluidics
, see https://spherefluidics.com/our-products/platform-products/cyto-mine-chroma/ for “Cyto-Mine Chroma” (last accessed April 10, 2024).
156.
Sphere Fluidics
, see https://spherefluidics.com/our-products/platform-products/cyto-mine/ for “Cyto-Mine®: Accelerating Antibody Discovery and Cell Line Development” (last accessed April 10, 2024).
157.
A.
Zinchenko
,
S. R. A.
Devenish
,
B.
Kintses
,
P.-Y.
Colin
,
M.
Fischlechner
, and
F.
Hollfelder
, “
One in a million: Flow cytometric sorting of single cell-lysate assays in monodisperse picolitre double emulsion droplets for directed evolution
,”
Anal. Chem.
86
(
5
),
2526
2533
(
2014
).
158.
S.
Okushima
,
T.
Nisisako
,
T.
Torii
, and
T.
Higuchi
, “
Controlled production of monodisperse double emulsions by two-step droplet breakup in microfluidic devices
,”
Langmuir
20
(
23
),
9905
9908
(
2004
).
159.
A. S.
Utada
,
E.
Lorenceau
,
D. R.
Link
,
P. D.
Kaplan
,
H. A.
Stone
, and
D. A.
Weitz
, “
Monodisperse double emulsions generated from a microcapillary device
,”
Science
308
,
537
541
(
2005
).
160.
L. L. A.
Adams
,
T. E.
Kodger
,
S.-H.
Kim
,
H. C.
Shum
,
T.
Franke
, and
D. A.
Weitz
, “
Single step emulsification for the generation of multi-component double emulsions
,”
Soft Matter
8
,
10719
10724
(
2012
).
161.
K. K.
Brower
,
K. K.
Brower
,
C.
Carswell-Crumpton
,
S.
Klemm
,
B.
Cruz
,
G.
Kim
,
S. G. K.
Calhoun
,
L.
Nichols
,
P. M.
Fordyce
,
P. M.
Fordyce
,
P. M.
Fordyce
, and
P. M.
Fordyce
, “
Double emulsion flow cytometry with high-throughput single droplet isolation and nucleic acid recovery
,”
Lab Chip
20
(
12
),
2062
2074
(
2020
).
162.
Bio-Techne
, see https://www.bio-techne.com/brands/namocell for “Namocell Single Cell Dispensers Join Bio-Techne - The Fastest Way to Single Cell Isolation” (last accessed April 10, 2024).
163.
K. K.
Brower
,
M.
Khariton
,
P. H.
Suzuki
,
C.
Still
,
G.
Kim
,
S. G. K.
Calhoun
,
L. S.
Qi
,
B.
Wang
, and
P. M.
Fordyce
, “
Double emulsion picoreactors for high-throughput single-cell encapsulation and phenotyping via FACS
,”
Anal. Chem.
92
,
13262
13270
(
2020
).
164.
T.
Cowell
and
H. S.
Han
, “
Double emulsion flow cytometry for rapid single genome detection
,”
Methods Mol. Biol.
2689
,
155
167
(
2023
).
165.
D. J.
Sukovich
,
S. T.
Lance
, and
A. R.
Abate
, “
Sequence specific sorting of DNA molecules with FACS using 3dPCR
,”
Sci. Rep.
7
,
39385
(
2017
).
166.
S.
Zhuang
,
L.
Semenec
,
S. S.
Nagy
,
A. K.
Cain
, and
D. W.
Inglis
, “
High-precision screening and sorting of double emulsion droplets
,”
Cytometry, Part A
105
(
7
),
547
554
(
2024
).
167.
Y.
Jia
,
Y.
Ren
,
L.
Hou
,
W.
Liu
,
T.
Jiang
,
X.
Deng
,
Y.
Tao
, and
H.
Jiang
, “
Electrically controlled rapid release of actives encapsulated in double-emulsion droplets
,”
Lab Chip
18
(
7
),
1121
1129
(
2018
).
168.
Y.
Jia
,
Y.
Ren
,
W.
Liu
,
L.
Hou
,
Y.
Tao
,
Q.
Hu
, and
H.
Jiang
, “
Electrocoalescence of paired droplets encapsulated in double-emulsion drops
,”
Lab Chip
16
(
22
),
4313
4318
(
2016
).
169.
L.
Hou
,
Y.
Ren
,
Y.
Jia
,
X.
Deng
,
W.
Liu
,
X.
Feng
, and
H.
Jiang
, “
Continuously electrotriggered core coalescence of double-emulsion drops for microreactions
,”
ACS Appl. Mater. Interfaces
9
(
14
),
12282
12289
(
2017
).
170.
A.
Stucki
,
P.
Jusková
,
N.
Nuti
,
S.
Schmitt
, and
P. S.
Dittrich
, “
Synchronized reagent delivery in double emulsions for triggering chemical reactions and gene expression
,”
Small Methods
5
(
8
),
2100331
(
2021
).
171.
Y.
Kee Jo
,
D.
Lee
,
Y. K.
Jo
, and
D.
Lee
, “
Biopolymer microparticles prepared by microfluidics for biomedical applications
,”
Small
16
,
1903736
(
2020
).
172.
G.
Leonaviciene
,
K.
Leonavicius
,
R.
Meskys
, and
L.
Mazutis
, “
Multi-step processing of single cells using semi-permeable capsules
,”
Lab Chip
20
(
21
),
4052
4062
(
2020
).
173.
S.
Ma
,
J.
Thiele
,
X.
Liu
,
Y.
Bai
,
C.
Abell
, and
W. T. S.
Huck
, “
Fabrication of microgel particles with complex shape via selective polymerization of aqueous two-phase systems
,”
Small
8
(
15
),
2356
2360
(
2012
).
174.
Q.
Liu
,
M.
Zhao
,
S.
Mytnyk
,
B.
Klemm
,
K.
Zhang
,
Y.
Wang
,
D.
Yan
,
E.
Mendes
, and
J. H.
van Esch
, “
Self-orienting hydrogel micro-buckets as novel cell carriers
,”
Angew. Chem., Int. Ed.
58
(
2
),
547
551
(
2019
).
175.
J.
De Rutte
,
R.
Dimatteo
,
M. M.
Archang
,
M.
Van Zee
,
D.
Koo
,
S.
Lee
,
A. C.
Sharrow
,
P. J.
Krohl
,
M.
Mellody
,
S.
Zhu
,
J. V.
Eichenbaum
,
M.
Kizerwetter
,
S.
Udani
,
K.
Ha
,
R. C.
Willson
,
A. L.
Bertozzi
,
J. B.
Spangler
,
R.
Damoiseaux
, and
D.
Di Carlo
, “
Suspendable hydrogel nanovials for massively parallel single-cell functional analysis and sorting
,”
ACS Nano
16
,
7242
7257
(
2022
).
176.
J. M.
Duarte
,
I.
Barbier
, and
Y.
Schaerli
, “
Bacterial microcolonies in gel beads for high-throughput screening of libraries in synthetic biology
,”
ACS Synth. Biol.
6
(
11
),
1988
1995
(
2017
).
177.
Y.
Nishikawa
,
M.
Kogawa
,
M.
Hosokawa
,
R.
Wagatsuma
,
K.
Mineta
,
K.
Takahashi
,
K.
Ide
,
K.
Yura
,
H.
Behzad
,
T.
Gojobori
, and
H.
Takeyama
, “
Validation of the application of gel beads-based single-cell genome sequencing platform to soil and seawater
,”
ISME Commun.
2
(
1
),
92
(
2022
).
178.
M.
Fischlechner
,
Y.
Schaerli
,
M. F.
Mohamed
,
S.
Patil
,
C.
Abell
, and
F.
Hollfelder
, “
Evolution of enzyme catalysts caged in biomimetic gel-shell beads
,”
Nat. Chem.
6
(
9
),
791
796
(
2014
).
179.
G.
Leonaviciene
and
L.
Mazutis
, “
RNA cytometry of single-cells using semi-permeable microcapsules
,”
Nucleic Acids Res.
51
(
1
),
e2
(
2023
).
180.
K.
Alessandri
,
B. R.
Sarangi
,
V. V.
Gurchenkov
,
B.
Sinha
,
T. R.
Kießling
,
L.
Fetler
,
F.
Rico
,
S.
Scheuring
,
C.
Lamaze
,
A.
Simon
,
S.
Geraldo
,
D.
Vignjević
,
H.
Doméjean
,
L.
Rolland
,
A.
Funfak
,
J.
Bibette
,
N.
Bremond
, and
P.
Nassoy
, “
Cellular capsules as a tool for multicellular spheroid production and for investigating the mechanics of tumor progression in vitro
,”
Proc. Natl. Acad. Sci. U.S.A.
110
(
37
),
14843
14848
(
2013
).
181.
R.
Ghosh
,
A.
Arnheim
,
M.
van Zee
,
L.
Shang
,
C.
Soemardy
,
R. C.
Tang
,
M.
Mellody
,
S.
Baghdasarian
,
E.
Sanchez Ochoa
,
S.
Ye
,
S.
Chen
,
C.
Williamson
,
A.
Karunaratne
, and
D.
Di Carlo
, “
Lab on a particle technologies
,”
Anal. Chem.
96
,
7817
7839
(
2024
).
182.
Z.
Zhu
,
W.
Zhang
,
X.
Leng
,
M.
Zhang
,
Z.
Guan
,
J.
Lu
, and
C. J.
Yang
, “
Highly sensitive and quantitative detection of rare pathogens through agarose droplet microfluidic emulsion PCR at the single-cell level
,”
Lab Chip
12
(
20
),
3907
3913
(
2012
).
183.
H.
Huang
,
Y.
Yu
,
Y.
Hu
,
X.
He
,
O.
Berk Usta
, and
M. L.
Yarmush
, “
Generation and manipulation of hydrogel microcapsules by droplet-based microfluidics for mammalian cell culture
,”
Lab Chip
17
(
11
),
1913
1932
(
2017
).
184.
E. Z.
Macosko
,
A.
Basu
,
R.
Satija
,
J.
Nemesh
,
K.
Shekhar
,
M.
Goldman
,
I.
Tirosh
,
A. R.
Bialas
,
N.
Kamitaki
,
E. M.
Martersteck
,
J. J.
Trombetta
,
D. A.
Weitz
,
J. R.
Sanes
,
A. K.
Shalek
,
A.
Regev
, and
S. A.
McCarroll
, “
Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets
,”
Cell
161
(
5
),
1202
1214
(
2015
).
185.
A. M.
Klein
,
L.
Mazutis
,
I.
Akartuna
,
N.
Tallapragada
,
A.
Veres
,
V.
Li
,
L.
Peshkin
,
D. A.
Weitz
, and
M. W.
Kirschner
, “
Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells
,”
Cell
161
(
5
),
1187
1201
(
2015
).
186.
F. V.
De Rop
,
J. N.
Ismail
,
C.
Bravo González-Blas
,
G. J.
Hulselmans
,
C. C.
Flerin
,
J.
Janssens
,
K.
Theunis
,
V. M.
Christiaens
,
J.
Wouters
,
G.
Marcassa
,
J.
De Wit
,
S.
Poovathingal
, and
S.
Aerts
, “
Hydrop enables droplet-based single-cell ATAC-seq and single-cell RNA-seq using dissolvable hydrogel beads
,”
eLife
11
,
e73971
(
2022
).
187.
Samplix
, see https://samplix.com/products for “Samplix products” (last accessed April 10, 2024).
188.
Atrandi Biosciences
, see https://atrandi.com/products/flux for “Flux” (last accessed April 10, 2024).
189.
Partillion Bioscience
, see https://www.partillion.com/technology for “Nanovial Technology at a Glance” (last accessed April 10, 2024).
190.
S. L.
Sjostrom
,
Y.
Bai
,
M.
Huang
,
Z.
Liu
,
J.
Nielsen
,
H. N.
Joensson
, and
H.
Andersson Svahn
, “
High-throughput screening for industrial enzyme production hosts by droplet microfluidics
,”
Lab Chip
14
(
4
),
806
813
(
2014
).
191.
J. D.
Schnettler
,
M. S.
Wang
,
M.
Gantz
,
H. A.
Bunzel
,
C.
Karas
,
F.
Hollfelder
, and
M. H.
Hecht
, “
Selection of a promiscuous minimalist cAMP phosphodiesterase from a library of de novo designed proteins
,”
Nat. Chem.
16
(
7
),
1200
1208
(
2024
).
192.
J.
Vallapurackal
,
A.
Stucki
,
A. D.
Liang
,
J.
Klehr
,
P. S.
Dittrich
, and
T. R.
Ward
, “
Ultrahigh-Throughput screening of an artificial metalloenzyme using double emulsions
,”
Angew. Chem., Int. Ed.
61
,
e202207328
(
2022
).
193.
M.
Ryckelynck
,
S.
Baudrey
,
C.
Rick
,
A.
Marin
,
F.
Coldren
,
E.
Westhof
, and
A. D.
Griffiths
, “
Using droplet-based microfluidics to improve the catalytic properties of RNA under multiple-turnover conditions
,”
RNA
21
(
3
),
458
469
(
2015
).
194.
J. M.
Holstein
,
C.
Gylstorff
, and
F.
Hollfelder
, “
Cell-free directed evolution of a protease in microdroplets at ultrahigh throughput
,”
ACS Synth. Biol.
10
(
2
),
252
257
(
2021
).
195.
F.
Lan
,
B.
Demaree
,
N.
Ahmed
, and
A. R.
Abate
, “
Single-cell genome sequencing at ultra-high-throughput with microfluidic droplet barcoding
,”
Nat. Biotechnol.
35
(
7
),
640
646
(
2017
).
196.
G. X. Y.
Zheng
,
J. M.
Terry
,
P.
Belgrader
,
P.
Ryvkin
,
Z. W.
Bent
,
R.
Wilson
,
S. B.
Ziraldo
,
T. D.
Wheeler
,
G. P.
McDermott
,
J.
Zhu
,
M. T.
Gregory
,
J.
Shuga
,
L.
Montesclaros
,
J. G.
Underwood
,
D. A.
Masquelier
,
S. Y.
Nishimura
,
M.
Schnall-Levin
,
P. W.
Wyatt
,
C. M.
Hindson
,
R.
Bharadwaj
,
A.
Wong
,
K. D.
Ness
,
L. W.
Beppu
,
H. J.
Deeg
,
C.
McFarland
,
K. R.
Loeb
,
W. J.
Valente
,
N. G.
Ericson
,
E. A.
Stevens
,
J. P.
Radich
,
T. S.
Mikkelsen
,
B. J.
Hindson
, and
J. H.
Bielas
, “
Massively parallel digital transcriptional profiling of single cells
,”
Nat. Commun.
8
,
14049
(
2017
).
197.
P.
Samanta
,
S. F.
Cooke
,
R.
McNulty
,
S.
Hormoz
, and
A.
Rosenthal
, “
ProBac-seq, a bacterial single-cell RNA sequencing methodology using droplet microfluidics and large oligonucleotide probe sets
,”
Nature Protocols
19
,
2939
2966
(
2024
).
198.
S.
Chen
,
B. B.
Lake
, and
K.
Zhang
, “
High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell
,”
Nat. Biotechnol.
37
(
12
),
1452
1457
(
2019
).
199.
F.
Salmen
,
J.
De Jonghe
,
T. S.
Kaminski
,
A.
Alemany
,
G. E.
Parada
,
J.
Verity-Legg
,
A.
Yanagida
,
T. N.
Kohler
,
N.
Battich
,
F.
van den Brekel
,
A. L.
Ellermann
,
A. M.
Arias
,
J.
Nichols
,
M.
Hemberg
,
F.
Hollfelder
, and
A.
van Oudenaarden
, “
High-throughput total RNA sequencing in single cells using VASA-seq
,”
Nat. Biotechnol.
40
(
12
),
1780
1793
(
2022
).
200.
J.
De Jonghe
,
T. S.
Kaminski
,
D. B.
Morse
,
M.
Tabaka
,
A. L.
Ellermann
,
T. N.
Kohler
,
G.
Amadei
,
C. E.
Handford
,
G. M.
Findlay
,
M.
Zernicka-Goetz
,
S. A.
Teichmann
, and
F.
Hollfelder
, “
Spindrop: A droplet microfluidic platform to maximise single-cell sequencing information content
,”
Nat. Commun.
14
(
1
),
4788
(
2023
).
201.
A.
Gérard
,
A.
Woolfe
,
G.
Mottet
,
M.
Reichen
,
C.
Castrillon
,
V.
Menrath
,
S.
Ellouze
,
A.
Poitou
,
R.
Doineau
,
L.
Briseno-Roa
,
P.
Canales-Herrerias
,
P.
Mary
,
G.
Rose
,
C.
Ortega
,
M.
Delincé
,
S.
Essono
,
B.
Jia
,
B.
Iannascoli
,
O.
Richard-Le Goff
,
R.
Kumar
,
S. N.
Stewart
,
Y.
Pousse
,
B.
Shen
,
K.
Grosselin
,
B.
Saudemont
,
A.
Sautel-Caillé
,
A.
Godina
,
S.
McNamara
,
K.
Eyer
,
G. A.
Millot
,
J.
Baudry
,
P.
England
,
C.
Nizak
,
A.
Jensen
,
A. D.
Griffiths
,
P.
Bruhns
, and
C.
Brenan
, “
High-throughput single-cell activity-based screening and sequencing of antibodies using droplet microfluidics
,”
Nat. Biotechnol.
38
(
6
),
715
721
(
2020
).
202.
J.
Rutkauskaite
,
S.
Berger
,
S.
Stavrakis
,
O.
Dressler
,
J.
Heyman
,
X.
Casadevall i Solvas
,
A.
deMello
, and
L.
Mazutis
, “
High-throughput single-cell antibody secretion quantification and enrichment using droplet microfluidics-based FRET assay
,”
IScience
25
(
7
),
104515
(
2022
).
203.
R.
Gaa
,
E.
Menang-Ndi
,
S.
Pratapa
,
C.
Nguyen
,
S.
Kumar
, and
A.
Doerner
, “
Versatile and rapid microfluidics-assisted antibody discovery
,”
MAbs
13
(
1
),
1978130
(
2021
).
204.
K.
Fischer
,
A.
Lulla
,
T. Y.
So
,
P.
Pereyra-Gerber
,
M. I. J.
Raybould
,
T. N.
Kohler
,
J. C.
Yam-Puc
,
T. S.
Kaminski
,
R.
Hughes
,
G. L.
Pyeatt
,
F.
Leiss-Maier
,
P.
Brear
,
N. J.
Matheson
,
C. M.
Deane
,
M.
Hyvönen
,
J. E. D.
Thaventhiran
, and
F.
Hollfelder
, “
Rapid discovery of monoclonal antibodies by microfluidics-enabled FACS of single pathogen-specific antibody-secreting cells
,”
Nat. Biotechnol.
(published online).
205.
Z.
Jiang
,
H.
Shi
,
X.
Tang
, and
J.
Qin
, “
Recent advances in droplet microfluidics for single-cell analysis
,”
TrAC, Trends Anal. Chem.
159
,
116932
(
2023
).
206.
K.
Ngocho
,
X.
Yang
,
Z.
Wang
,
C.
Hu
,
X.
Yang
,
H.
Shi
,
K.
Wang
, and
J.
Liu
, “
Synthetic cells from droplet-based microfluidics for biosensing and biomedical applications
,”
Small
20
(
33
),
2400086
(
2024
).
207.
Y.
Wang
,
M.
Liu
,
Y.
Zhang
,
H.
Liu
, and
L.
Han
, “
Recent methods of droplet microfluidics and their applications in spheroids and organoids
,”
Lab Chip
23
(
5
),
1080
1096
(
2023
).
208.
Z.
Chen
,
S.
Kheiri
,
E. W. K.
Young
, and
E.
Kumacheva
, “
Trends in droplet microfluidics: From droplet generation to biomedical applications
,”
Langmuir
38
(
20
),
6233
6248
(
2022
).
209.
Y. G.
Kim
,
S.
Park
, and
S. H.
Kim
, “
Designing photonic microparticles with droplet microfluidics
,”
Chem. Commun.
58
(
74
),
10303
10328
(
2022
).
210.
A.
Lashkaripour
,
C.
Rodriguez
,
N.
Mehdipour
,
R.
Mardian
,
D.
McIntyre
,
L.
Ortiz
,
J.
Campbell
, and
D.
Densmore
, “
Machine learning enables design automation of microfluidic flow-focusing droplet generation
,”
Nat. Commun.
12
(
1
),
25
(
2021
).