You are on page 1of 28

Lab on a Chip

View Article Online


TUTORIAL REVIEW View Journal | View Issue

Microfluidic-assisted fabrication of carriers for


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Cite this: Lab Chip, 2017, 17, 1856


controlled drug delivery
Dongfei Liu, *ab Hongbo Zhang,bc Flavia Fontana,a
Jouni T. Hirvonena and Hélder A. Santos *ad

The microfluidic technique has brought unique opportunities toward the full control over the production pro-
cesses for drug delivery carriers, owing to the miniaturisation of the fluidic environment. In comparison to
the conventional batch methods, the microfluidic setup provides a range of advantages, including the im-
proved controllability of material characteristics, as well as the precisely controlled release profiles of pay-
loads. This review gives an overview of different fluidic principles used in the literature to produce either
Received 6th March 2017, polymeric microparticles or nanoparticles, focusing on the materials that could have an impact on drug de-
Accepted 28th April 2017
livery. We also discuss the relations between the particle size and size distribution of the obtained carriers,
and the design and configuration of the microfluidic setups. Overall, the use of microfluidic technologies
DOI: 10.1039/c7lc00242d
brings exciting opportunities to expand the body of knowledge in the field of controlled drug delivery and
rsc.li/loc great potential to clinical translation of drug delivery systems.

1 Introduction
a Recently, there has been an explosion in research toward the
Division of Pharmaceutical Chemistry and Technology, Drug Research Program,
Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. biomedical applications of micro/nano-materials.1–3 Poly-
E-mail: dongfei.liu@helsinki.fi, helder.santos@helsinki.fi meric particles are being employed for imaging and drug de-
b
John A. Paulson School of Engineering and Applied Sciences, Harvard University, livery in several diseases treatment. For example, polymeric
Cambridge, Massachusetts 02138, USA particles have been used for theranostics of cancer,4,5 therapy
c
Department of Pharmaceutical Science, Åbo Akademi University, FI-20520 Turku,
of diabetes,6 and tissue engineering.7 In the innovative fields
Finland
d
Helsinki Institute of Life Science, HiLIFE, University of Helsinki, FI-00014 of immunotherapy (treatment of autoimmune diseases8 or
Helsinki, Finland immunostimulation for cancer therapy9,10) and endothelial

Dongfei Liu earned his Ph.D. in Dr. Hongbo Zhang graduated his
pharmacy at the University of PhD in December 2012 in Fac-
Helsinki in 2014. He is well- ulty of Pharmacy, University of
versed in a variety of fields, such Helsinki. He is specialized in
as nanomedicine, microfluidics, nanomedicine, imaging, con-
biomaterials, cancer theranostics, trolled drug delivery, micro-
and spinal cord injury therapy fluidics, molecular biology and
among others. Primarily, he is drug metabolism. Since January
interested in how to engineer bio- 2013, Dr. Zhang joined Associate
materials with ultrahigh mass Prof. Hélder A. Santos' group in
fraction of therapeutics for con- University of Helsinki as a Post-
trolled drug delivery. In addition, doc and then he joined Prof. Da-
Dongfei Liu he has received several competi- Hongbo Zhang vid A. Weitz's group in Harvard
tive grants and awards, such as University since March 2014 as
the Jane and Aatos Erkko Foundation (2016–2018), Research Funds a visiting scholar. In September 2016, he became a Tenure Track
of the University of Helsinki (2017–2020), and the Best Doctoral Assistant Professor in Department of Pharmaceutical Sciences
Dissertation of the University of Helsinki in 2014. In February Laboratory, Åbo Akademi University and established his group fo-
2017, he became a principal investigator at the Faculty of cused on “Functional Materials and Microfluidics”.
Pharmacy, University of Helsinki.

1856 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

diseases therapy, polymeric particles are playing an production techniques can be classified into top-down and
increasing role.11 The favorable characteristics of micro/nano bottom-up approaches.14 In the top-down technique, parti-
drug delivery systems derive from the modifications in the cles are usually prepared by the mechanical fragmentation
pharmacokinetic profiles of loaded therapeutics.2 Specifically, of bulk precursors. In contrast, monomers assemble into
the micro/nano drug delivery systems offer a protective effect a micro/nano-sized structure in the bottom-up approach.
against the degradation of therapeutics, controlled drug re- Alternatively, the particle synthesis approaches can be cat-
lease profiles, an enhanced dissolution rate of poorly water- egorized into types such as emulsion, precipitation and di-
soluble drugs, and the possibility of precisely targeting the rect compositing methods, based on the particle formation
drug molecules to the site of action.12 These beneficial fea- general principles.15 Toward the emulsion-based method,
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

tures of micro/nano drug delivery systems enable the use of a particle precursors are usually dissolved in a water immis-
lower drug dose with reduced side effects compared to the cible organic solvent and then emulsified by a larger vol-
bare drug molecules.13 ume of aqueous solution,15 forming simple, double or
Micro/nano polymeric particles are usually prepared by multiple emulsions. The polymeric micro/nano-particles
the conventional batch approach. The conventional particle are then obtained by solvent-extraction or evaporation.16
The precipitation-based method, such as nanoprecipitation,
salting out and dialysis,15 can also be employed to synthe-
size polymeric micro/nano-particles. In a common precipi-
Flavia Fontana (MSc. Pharm.) is tation approach, the particle precursors are dissolved in a
a graduate student in Associate water miscible organic solvent. This organic solution is
Professor Santos' lab, at the Fac- added to a liquid where the particle precursor is insolu-
ulty of Pharmacy, University of ble, leading to a polymer phase separation and subse-
Helsinki (Finland). She obtained quently forming polymeric particles.17 The third approach
her Master's Degree in Medicinal is the direct compositing method, such as spray drying,
Chemistry and Pharmaceutical supercritical fluid, melting technique, and in situ forming
Technology at the University of polymeric particles.15
Pavia (Italy). Her main research Translation of basic research into clinical applications
interests are the development of is defined as translational medicine.18 Despite of their ad-
innovative nanovaccines, immu- vantages over the bare drug molecules, the clinical transla-
notherapy, and microfluidics. tion of polymeric drug delivery systems is relatively slow.
Flavia Fontana She has published papers in The slow translation rate can be partially ascribed to the
journals like Advanced Materials poorly control of the preparation processes in the conven-
and ACS Applied Materials and Interfaces. tional batch method. For example, the polymeric particles
prepared by the conventional batch method usually show

Professor Hirvonen is the PI of Associate Professor Hélder A.


about 25 externally funded re- Santos (D.Sc. Tech., Chem. Eng.)
search projects. He has super- is the Head of the Research Unit
vised more than 25 PhD theses Pharmaceutical Nanotechnology
and co-authored more than 200 and Chemical Microsystems, the
peer reviewed research articles Head of the Preclinical Drug For-
and patents. He is an expert in mulation and Analysis Group at
the research and development of the Faculty of Pharmacy, Univer-
novel biomaterials, pharmaceuti- sity of Helsinki. His current work
cal technology applications, and bridges between medical engi-
controlled/targeted nanotechnol- neering and pharmaceutics, fo-
ogy approaches for drug delivery cusing on nanomedicines and
Jouni T. Hirvonen systems. In 2007, he received the Hélder A. Santos microfluidics technology for con-
CRS/Eurand Grand Prize Award trolled drug delivery and nano-
based on novel approaches for oral drug delivery applications. He particle fabrication. He has authored over 190 publications. He
acts as a pharmaceutics expert in the national and international received the “Talent Prize in Science” (Portugal) in 2010, the Euro-
drug regulation development. He is an Executive Committee mem- pean Research Council Starting Grant in 2013, the Young Re-
ber in the European Association of Faculties of Pharmacy. searcher Award in 2013 (Finland), and the Academy of Finland
Award in 2016.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1857
View Article Online

Tutorial review Lab on a Chip

high batch-to-batch variations in physicochemical proper- 2 Microfluidic devices and


ties, such as the average particle size, size distribution, technologies for drug delivery
surface charge and drug release profiles.19–21 To precisely
control the release of payloads, it is critical to prepare the 2.1 Materials for microfluidic devices
polymeric particles with known sizes and size distribu- The microfluidic devices can be fabricated in a variety of dif-
tions, since the particle size strongly affect the payload re- ferent materials, mostly from polydimethylsiloxane, glass, sil-
lease rate.22 By tailoring their internal structures, the load- icon, fused silica, hydrogel, and polytetrafluoroethylene.31–34
ing of therapeutics in polymeric particles and the release Polydimethylsiloxane devices are usually fabricated by soft-
of payloads can also be tuned.23 For example, co-delivery lithography.35 In a general process, polydimethylsiloxane and
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

of multiple drugs can be attained through adjusting the the curing agent are poured onto a master and cured; the
size and number of internal compartments.24 Another polymer replica is then detached from the master and, revers-
strategy to manipulate the release of payloads is using ibly or irreversibly, attached to a second layer to seal the
stimulus-responsive materials to synthesize the polymeric channels.35 An alternative approach to produce polydimethyl-
particles.25 After exposure to the environmental triggers, siloxane devices is 3D printing; however, the printing resolu-
such as pH, temperature or ionic strength, the particles tion is still lower than the conventional photolithogra-
undergo a physicochemical change and then release the phy.32,36 Polydimethylsiloxane structures can serve as molds
payloads.26 for the hot embossing of films made of fluorinated poly-
Microfluidics has been defined as the manipulation of mers.34 Polydimethylsiloxane devices can also be fabricated
fluids in channels with dimension of tens of microme- by using glass capillaries templated agarose molds, which are
ters.27 Microfluidics has been extensively applied in the removed by dissolving them with boiling water.37 Micro-
fabrication of micro/nano-materials with precisely controlled fluidic devices prepared by laser direct-writing of thermoplas-
physicochemical features.28 Due to its excellent ability to tic materials (e.g., polystyrene) showed lower cost and less
manipulate nanoliter flows,27,29,30 microfluidics has fabrication time than the conventional hot embossing and
emerged as an alternative technique to the conventional photolithography.33
methods for the preparation of micro/nano-particles. In Glass capillary microfluidic devices are usually fabri-
comparison to the conventional methods, the particle cated by pulling capillaries into a fine tip with an orifice
preparation process is miniaturized in the microfluidic de- of precise size, co-axially assembly within a larger square
vice, thereby leading to a reduction in the consumption of or round capillary, and lastly gluing those capillaries onto
reagents. Furthermore, microfluidic approaches enable the a glass slide.38 According to the potential applications of
continuous online synthesis of polymeric particles, which the device, the capillaries can be assembled in different
could reduce the batch-to-batch variations in the physico- combinations; moreover, the inner and outer surfaces of
chemical properties of the obtained particles.19 these capillaries can be modified into hydrophobic or hy-
The favorable features of the microfluidic technique are drophilic by simple chemical reactions.28,38 Channels
summarized in Fig. 1. In this review, we first highlight the formed by glass etching is an alternatively strategy to pre-
use of droplet microfluidics to produce polymeric microparti- pare glass microfluidic devices, which are technically de-
cles and capsules for controlled drug loading and release, manding and time consuming.39 A reusable and user-
and then summarize the recent advances of microfluidic friendliness approach to facilitate the fabrication of micro-
techniques toward the preparation of polymeric nanoparticles capillary devices was recently proposed.40 In this approach,
regarding drug delivery. the capillaries are assembled rapidly into a metallic struc-
ture, avoiding the advanced skills and reducing the assem-
bly from hours to minutes, in comparison to the conven-
tional preparation approach for glass capillary devices.
Polydimethylsiloxane is easier to be tailored to a variety of
channel geometries,23,41 ranging from the co-axial align-
ment of two channels to the complexed multicompartment
systems.41,42 However, polydimethylsiloxane dissolves in, or
is swelled by, many common organic solvents.43 The low
elastic modulus of polydimethylsiloxane is a critical issue
for high pressure and high flow rate operation, because
this leads to a large alteration of channel geometry.44
Glass capillaries can better tolerate a wider range of sol-
vents and they are more flexible regarding applications in
drug encapsulation. Additionally, glass is a material of
choice in fabricating microfluidic devices due to its excel-
Fig. 1 Favorable features of microfluidic techniques toward the lent resistance to high pressures and flow rates.39 Each
preparation of polymeric particles. material has its own features regarding the easiness and

1858 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

cost of the device fabrication, the withstanding of high ily optimized, showing the high versatility and strong robust-
pressures and flow rates, as well as the resistance to or- ness of the microfluidic platform.52 The droplet microfluidics
ganic solvents.45 and microfluidic mixing is discussed in detail in sections 3
and 4, respectively.

2.2 Microfluidic technologies for micro/nano-particles


preparation 2.3 Effect of the physicochemical properties of drug delivery
systems on their interaction with biological systems
Droplet-based microfluidics is an important subcategory of
the microfluidic technologies, which manipulates immiscible The physicochemical properties of drug delivery systems af-
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

phases to generate droplets.38 The droplet phase is identified fect their behavior in vitro and in vivo. As listed in Table 1,
as dispersed phase, while the medium phase is termed as the shape, size, stiffness, and surface charge and
continuous phase. In general, the droplet size is controlled functionalization are the most important factors, which affect
by adjusting the flow rates and ratios of the two phases; how- the uptake of carriers by cancer cells and the mononuclear
ever, it is also affected by the viscosity of the dispersed phase, phagocyte system, the circulation time of micro/nano-parti-
the channel and orifice diameter, flow regimes, and other pa- cles and, ultimately, their biodistribution.19,53 Toward a bet-
rameters.46 Droplet microfluidics is a versatile technique to ter control of the interactions between the drug delivery sys-
produce micro-sized single and multiple emulsions, because tems and the biological systems, accurate modulation of the
of its ability to produce droplets with various degrees of com- carrier properties is a prerequisite. In comparison to conven-
plexity.28,42,46 Droplet-based microfluidics techniques are tional bulk methods, microfluidic synthesis can fabricate
widely used in, for example, particle fabrication, encapsula- micro/nano-particles with varying properties in a well-
tion, sorting, and cell analysis.38,47,48 It represents also a very controlled manner, thus offering a convenient way for study-
useful technique for the fabrication of advanced drug delivery ing the interactions between micro/nano-particles and biolog-
systems with precisely controlled loading and release of ical systems, as well as the therapeutic efficacy of micro/
therapeutics.48 nano-particles.
Benefiting from the small channel dimension and the
resulting large surface-to-volume ratio, the microfluidic de- 3 Droplet microfluidics toward
vices can drastically reduce the mixing path of solvent and controlled drug delivery
non-solvent (miscible phases) down to few tens of microme-
ters for the preparation of nanoparticles. The short mixing 3.1 Flow regimes in microfluidic channels
path facilitates the fast mixing process, resulting in a control- Microfluidic technology significantly reduces materials cost
lable and tunable mixing time from the millisecond to micro- and enhances the drug encapsulation efficiency.23 In addi-
second scale.49 The quick assembly or the precipitation of tion, the droplets formed in microfluidics present high
particle precursors results in the smaller particle size and monodispersity of size and can be adjusted by exploiting the
narrower size distribution, in comparison to the conventional physical properties of the fluids. Reynolds number (Re) and
methods.19,50,51 By simply varying the flow rates, polymer Péclet number (Pe) are two dimensionless parameters that re-
compositions and polymer concentrations, the physico- flect the characteristics of the fluid flow and the action of
chemical properties of the obtained nanoparticles can be eas- molecules within the fluid.70 Reynolds number can be

Table 1 The physicochemical properties of the drug delivery systems affecting their in vitro and in vivo behaviours

Properties Interaction with cells In vivo behavior


Shape Particles with larger aspect ratios and sharper angular are Tumor accumulation of nanospheres and nanodisks (on the
taken up in larger amounts and faster rates;54,55 cylindrical surface of tumor) is higher than that of nanorods and
particles internalize at a higher percentage than cubic ones56 nanocages (throughout the tumors);57 the circulation time of
filomicelles (ca. one week) was ten times longer than their
spherical counterparts58
Size Upper limit of particle internalization into nonphagocytotic One of the most significant parameters on crossing the
cells is ca. 3 μm;56 phagocytosis decreases with increasing biological barriers and the biodistribution of particles;61
particle size;59 non-phagocytic cells favor the uptake of threshold of 200 nm for long circulating;62–64 effect on the
smaller particles compared to phagocytic cells60 efficient delivery of the payload61
65–67
Stiffness Stiff particles showing higher cell uptake Elastic particles have longer circulation time; stiff
nanoparticles preferentially found in the tumor site65–67
Surface charge Positively-charged particles interact more with the cells, Different complement activation pathways based on the
increasing the cellular uptake of particles without charge of the particle;62 unwanted liver uptake for positively
53
specification charged particles;63 effect on the delivery of payloads in
healthy animal models61
Surface Surface modification with targeting ligands, such as antibody Tumor accumulation of actively targeted nanoparticles (e.g.,
functionalization of vascular endothelial growth factor receptors,68 can antibody coated) is faster and higher relative to their passive
increase cellular uptake to specific cells counterparts [e.g., polyIJethylene glycol) (PEG)-coated]69

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1859
View Article Online

Tutorial review Lab on a Chip

calculated by eqn (1).

(1)

where, ρ is the density of the fluid, ν is the velocity of the


fluid, L is the characteristic liner dimension, and μ is the vis-
cosity of the fluid.70 The fluid flow with high Reynolds num-
ber (usually > 2300) is usually classified as turbulent flow,
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

presenting a chaotic pattern with no distinct streamlines Fig. 2 Microfluidic flow patterns and flow regimes. (a and b)
Schematic illustration of turbulent (a) and laminar (b) flows. (c and b)
(Fig. 2a).71 In contrast, the laminar flow reflects a flow pattern Schematic illustration of dripping (c) and jetting (d) regime.
with distinct streamlines which are parallel to the fluid direc-
tion (Fig. 2b). The onset of transition between the turbulent
and laminar flow occurs at Reynolds numbers of 1800–2000.
The laminar flow usually occurs at a low Re number we can control the droplet formation by adjusting the flow
(<1800).71 Since the laminar flow is characterized by steady rate, fluid viscosity, channel parameters, and other
streamlines, the fluids and the molecules within the fluids parameters.74
can be precisely manipulated to generate controllable and
monodisperse droplets, which represents the favorable fea-
tures of droplet microfluidics for drug encapsulation. 3.2 Device geometries of droplet microfluidics
The Péclet number is another important parameter in A single emulsion is the emulsification of one fluid in an-
microfluidics, which reflects the diffusion or convection of other immiscible fluid. The typical microfluidic single
molecules in the fluids.72 The Péclet number can be calcu- emulsion includes oil-in-water (O/W) and water-in-oil (W/O)
lated by eqn (2). emulsions. All three geometries, co-flow, flow-focusing and
T-junction, can be applied in single emulsion preparation
(Fig. 3a–c). The co-flow geometry defines a configuration
(2)
where the dispersed phase in inner capillary flows through
an orifice of defined dimension into a continuous flow from
where, u is the local velocity of the fluid, l is the characteris- outer capillary flowing in the same direction (Fig. 3a). In this
tic length, and D is the mass diffusion coefficient. In droplet configuration, the droplets are mainly produced due to
microfluidics, due to the small volumes and the laminar flow Rayleigh–Plateau instability, therefore the jetting regime is
pattern, the molecules transfer is slow, mainly through diffu- often involved.46 In flow-focusing droplet microfluidic de-
sion instead of convection. Therefore, microfluidics mini- vices, the dispersed phase and continuous phase flow
mizes the transfer of molecules from the dispersed phase to through two sides of the channel and meets before the ori-
the continuous phase, enabling the high encapsulation effi- fice of the inner capillary, while the droplets are formed at
ciency of drugs into the droplets. the orifice (Fig. 3b).46 The physical mechanisms involved in
The most popular geometries of droplet microfluidics are the flow-focusing geometry are complex, and both dripping
co-flow, flow-focusing, and T-junction, under a dripping or and jetting regimes can be formed in a device presenting
jetting regimen (Fig. 2c and d).23 The shape of the junction such geometry. In T-junction microfluidic devices (Fig. 3c),
helps to define the interface between the two immiscible the continuous phase flows through a straight main channel,
flows. The formation of a droplet is achieved through the cre- while the dispersed phase flows through side channels, enter-
ation of a dragging force higher than the viscosity force. ing the main channel through cross flow.75 T-junction has
When the free surface instabilities between the two phases the benefit of producing droplets with better size mono-
are large enough, the droplets form and break off from the dispersity. All three geometries and combination of geome-
dispersed phase; while the large volume of continuous phase tries (for the development of double and multiple emulsions)
stabilizes the droplets preventing them from merging with have been applied in producing microparticles for drug
each other. delivery.23,41
In dripping regime (Fig. 2c), the drop formation results Double emulsions are generally produced using a combi-
from a balance between the surface tension forces and the nation of two geometries (Fig. 3d–g). A common double
pulling viscous drag.73 In the case of jetting (Fig. 2d), the emulsion microfluidic device consists of two round capil-
droplet formation can be explained by Rayleigh–Plateau in- laries with the orifices facing each other; both round capil-
stability, a stream of fluid breaks up into smaller packets laries are inserted into a square capillary (Fig. 3d). The inner
with less surface area but the same volume.73 In this case, the phase flows through the inner capillary; the middle and outer
drop pinch-off is driven by the balance between the surface phases flow through the outer square capillary in the same
tension that pulls the fluid away and the viscous drag of the and opposite direction of the inner flow, respectively. There-
fluid that resists the flow. By understanding these theories, fore, this can be considered as a co-flow combining flow-

1860 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 3 Device geometries of droplet microfluidics. (a–c) The geometries, co-flow (a), flow focusing (b) and T-junction (c), of microfluidic devices
for the preparation of single emulsions. (d–g) Combined geometries to fabricate multiple emulsions, including co-flow combing flow focusing with
one inner fluid (d) and two inner fluids (e), sequential co-flows for thin shell capsule production (f), and sequential T-junction (g). (h) The device ge-
ometry for producing Janus particles with two parallel inner flows.

focusing geometry. Double emulsion is formed when the from droplet microfluidics (Fig. 3h). Janus particles are usu-
three fluids enter the inner capillary facing to the other inner ally synthesized by solidifying Janus droplets with the help of
inlet capillary, which acts as collection tube. Double emul- phase separation, polymerization or other processes.79 The
sion includes water-in-oil-in-water (W/O/W) and oil-in-water- fabrication of Janus particles originated in 2006,80 when re-
in-oil (O/W/O) systems. W/O/W emulsion has been widely searchers from the University of Tokyo reported the produc-
used in the development of drug delivery microcapsules with tion of droplets containing two hemispheres using a micro-
hollow core and a polymeric shell. By adjusting the flow rates fluidic device with the T-junction geometry (Fig. 3h). By
and ratios of the inner, middle, and continuous phases, the introducing nanoparticles that can be aggregated on one side
size of the inner and outer droplets can be controlled, thus of the droplets, hierarchal Janus structures can be simply pre-
manipulating the size and shell thickness of the produced pared by the flow focusing geometry for the preparation of
microcapsules.76 In dripping mode, the size of the double single emulsion.81 Because the formation of Janus droplets
emulsion drops is mainly governed through the flow rate of only takes place after the droplet formation, stability issues
continuous phase, while the thickness of the shell is deter- associated with the convective cross-flow of the monomer
mined by the middle layer (content between inner and outer fluids are avoided. We summarize the physicochemical fea-
phases). Thus, for a fixed flow rate of continuous phase, the tures, such as the average particle size and the release pro-
higher the flow ratio between the inner and middle phases, files, of the micro/nano-particles synthesized by droplet
the thicker the capsule shell.76 By simultaneously introducing microfluidics in Table 2; and discussed in detail in sections
two inner fluids (Fig. 3e), the multiple-component emulsions 3.3 and 3.4.
can be prepared.77 To obtain very thin shells, another
microfluidic double emulsion geometry has been developed
(Fig. 3f): the inner capillary is inserted into a middle capillary 3.3 Microcarriers synthesized by droplet microfluidics for
and then the middle capillary is inserted into the square controlled drug delivery
outer capillary facing the orifice of the collection capillary in- The O/W emulsion usually templates microparticles com-
side the outer capillary.76 In this case, the inner, middle and posed of hydrophobic materials. Many drug delivery plat-
outer phases flow through different capillaries, but in the forms are developed through O/W single emulsion, where
same direction, combining co-flow and co-flow geometry. the drugs are encapsulated into a biocompatible polymeric
Double emulsion can also be produced by T-junction geome- particle matrix, including poly(lactic-co-glycolic acid)
try by a two-stage process (Fig. 3g). In the first stage, the in- (PLGA), poly-ε-caprolactone and hydroxypropyl methylcellu-
ner fluid is encapsulated by the middle fluid, forming a sin- lose acetate succinate (HPMCAS).82,83 PLGA is a biodegrad-
gle emulsion. The droplets then flow into the second drop able and biocompatible copolymer that is used in a vari-
maker and become encapsulated by the outer fluid, forming ety of Food and Drug Administration approved therapeutic
the double emulsions. devices.16 The polymers used in O/W emulsions are typi-
Janus particles are special types of particles which com- cally dissolved in a volatile solvent which can evaporate or
bine two or more distinct materials at the surfaces;78 for ex- diffuse out from the droplets. The size of microparticles
ample, a particle presents one half hydrophobic surface and is primarily controlled by the droplet size and how much
the rest half hydrophilic. These particles can also be made the final droplet shrinks during solvent removal, an

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1861
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Table 2 Droplet based microfluidics for drug delivery applications

Average
Microfluidic devices Emulsion types Particle precursors Drug loading Dispersed and continuous phases size Estimated t50 and t100 Ref.
Tutorial review

Polydimethylsiloxane O/W PLGA Bupivacaine, ca. 20% Dichloromethane/water ca. 11 and t50 ≈ 25 days for 11 μm; >35 85
41 μm days for 41 μm
Polydimethylsiloxane O/W AcDX Sorafenib (SFN) and Dimethyl carbonate/2% poloxamer 407 4.1–6.8 μm Slow release at pH 7.4; burst 84
celecoxib, ca. 0.5–4.2% release at pH 2.5 with t100 < 30
min
Polydimethylsiloxane O/W HPMCAS Celecoxib, SFN, ca. Dimethyl carbonate/2% poloxamer 407 4.2 μm At pH 5.0, t50 ≈ 20 min, t100 ≈ 84
2.6% 60 min
Glass capillaries O/W Sodium alginate Iopamidol, ca. 17% Water/n-decanol with 5 wt% span 80 ca. 100 μm t50 ≈ 15–30 min and t100 ≈ 86

1862 | Lab Chip, 2017, 17, 1856–1883


60–120 min
Glass capillaries W/O/W Thermal sensitive lipid Doxorubicin (DOX) and Water/molten lipid/10% polyIJvinyl ca. 50–100 t100 ≈ 160 s at 37 °C 87
(paclitaxel) PTX alcohol) μm
Glass capillaries W/O/W Polyethylene glycol Bovine serum albumin 20% PEG 6000/ethyl acetate/5% ca. 39 μm t50 > 170 h 88
(PEG)/poly-ε-caprolactone polyIJvinyl alcohol)
Polydimethylsiloxane O/W/O Polyacrylamide/PNIPA Rhodamine B PNIPA microgel/PNIPA ca. 50 μm t50 ≈ 35 s when temperature < 89
isothiocyanate–dextran, solution/fluorocarbon oil 33 °C

Glass capillaries O/W PSi in HPMCAS mixtures Atorvastatin and Ethyl acetate/2% poloxamer 407 ca. 129 μm No drug released at pH 1.2; 83
celecoxib, ca. 15% for 40% released at pH 6.0; the
each rest released at pH 7.4
Glass capillaries O/W PSi in HPMCAS Fluorouracil and Ethyl acetate/2% poloxamer 407 ca. 30 μm No drug release at pH < 6.5; 90
celecoxib, ca. 7% the drugs are burst released at
pH 7.4
Glass capillaries O/W PSi in HPMCAS Glucagon-like Ethyl acetate/2% poloxamer 407 ca. 60 μm At pH 1.2, no drugs released; 24
peptide-1, 15% in PSi at pH 6.8, t50 ≈ 400 min
Glass capillaries O/W PSi in HPMCAS Dipeptidyl peptidase 4, Ethyl acetate/2% poloxamer 407 ca. 60 μm At pH 1.2, t50 < 10 min; at pH 24
— 6.8, released in minutes
Glass capillaries O/W Halloysite nanotubes in Atorvastatin, 2%; Ethyl acetate/2% poloxamer 407 ca. 70 μm No release at pH < 6.5; at pH 91
HPMCAS celecoxib, 6% 7.4, t50 and t100 < 30 min
Glass capillaries W/O/W PSi in phospholipid Piroxicam, 19% 8% PEG 6000 and 2% polyIJvinyl ca. 114 μm At pH 6.0, t100 ≈ 50 min; at pH 92
vesicle alcohol)/chloroform and hexane (1 : 1.18, 7.4, t100 ≈ 350 min
v/v)/10% polyIJvinyl alcohol)
Glass capillaries W/O/W PSi, DNA, gold nanorods DOX, 30%; erlotinib, Water/lipid in oil/10% polyIJvinyl ca. 100 μm t50 is ca. 9 h and t100 ≥ 24 h 93
in lipid 10%; 17-AAG, 15% alcohol)
Glass capillaries Janus particles PolyIJacrylamide)/ Ketoprofen and Acrylamide and methylacrylate ca. 59–240 t100 varies between 10–24 h 94
polyIJmethyl acrylate) sodium fluorescein solution/silicon oil μm with different morphologies
Polydimethylsiloxane W/O Alginate No drugs loaded Alginate solution/dimethyl carbonate ca. 1–50 — 95
μm and
10–300 nm
Polydimethylsiloxane O/W or O/O PLGA No drugs loaded Dimethyl carbonate/water ca. 70 — 96
nm–30 μm
Polydimethylsiloxane Water/air; Amorphous drug Fenofibrate, Drug solution/air ca. 14 nm — 97
ethanol/air nanoparticles clotrimazole, danazol
or estradiol
Polymethylmethacrylate Nitrogen Ethyl cellulose Amoxicillin, ca. 13% Ethanol/2% Tween 20 ca. 510 nm 65% (510 nm) or 87% (92 nm) 98
gas/ethanol/water or 90 nm of amoxicillin released in 12 h
Glass capillaries Fiber breakup to — No drugs loaded Polymer cladding 20 nm to 2 — 99
droplets (polyethersulphone)/chalcogenide glass mm
View Article Online

This journal is © The Royal Society of Chemistry 2017


Lab on a Chip
View Article Online

Lab on a Chip Tutorial review

adjustable variable by employing different polymer concen- Regardless of the morphologies of the microgels, the t50
trations.84 A central challenge in the preparation of drug- of iopamidol <25 min for the obtained alginate micro-
loaded PLGA microparticles is the poor control of emulsi- gels,86 which was much faster than that of PLGA micro-
fication processes, resulting in variable physicochemical particles (Fig. 4b).
properties and burst release profiles for the obtained par- Single emulsion does not ensure the simultaneous loading
ticles. With microfluidic method, the drug encapsulation of multiple therapeutics, even more difficult, when the pay-
is in general tight and the drug release is control by the loads present different solubility. Thereby, double emulsions
degradation or dissolution of the polymer matrix.85 are also widely used in drug delivery applications. Weitz's
PLGA and the model drug bupivacaine in group developed a generic, solvent-free one-step microfluidic
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

dichloromethane were flow focused by 1% polyIJvinyl double emulsion approach to simultaneously encapsulate a
alcohol) to form the single emulsion droplets by micro- hydrophilic and a hydrophobic drug with high efficiency.87
fluidics (Fig. 4a).85 Even with the same particle size, the In W/O/W double emulsion, doxorubicin (DOX) and paclitaxel
release profiles of bupivacaine from PLGA microparticles (PTX) were co-loaded into inner aqueous core and lipid shell,
prepared by droplet microfluidics was much slower than respectively, of the thermal sensitive lipid capsules (Fig. 4f).
those synthesized by the conventional method. Regarding The release of DOX and PTX was both controlled by the melt-
the PLGA microparticles prepared by the droplet micro- ing of lipid shell at 37 °C, ensuring the release of each pay-
fluidics, the release half-life (t50) of bupivacaine increased load at the same location (Fig. 4g). Moreover, Pessi et al.88
from ca. 26 days to >30 days by increasing the particle have encapsulated the water soluble bovine serum albumin
size from 11 to 41 μm (Fig. 4b). Vasiliauskas and Santos into poly-ε-caprolactone microcapsules. After encapsulation
et al.84 produced hollow microparticles, including PLGA, into the poly-ε-caprolactone capsules, the t50 of bovine serum
acetalated dextran (AcDX) and HPMCAS, by simply albumin was sustained from <1 min to >170 h. Moreover,
adjusting the speed of solvent removal of single emul- hollow microgels can be produced by microfluidic O/W/O
sions. Two model drugs, sorafenib (SFN) and celecoxib, emulsion. By adjusting the flow rates of inner, middle and
was simultaneously encapsulated into the hollow AcDX outer flows, the number of inner drops and the shell thick-
and HPMCAS microparticles, presenting similar drug re- ness of microgels can be adjusted (Fig. 4h).89 The release of
lease profiles. Concerning the AcDX microparticles, the Rhodamine B isothiocyanate-labeled dextran was controlled
higher the concentration of particle precursors (10–100 mg by the reswelling of the microgel shell as well as by the struc-
mL−1), the lower the loading degree of the payloads (4.2– ture of the microgel (Fig. 4i).
0.5%) inside the microparticles and the slower the release Microfluidics also represents an effective technique for
rate of payloads. Regarding the AcDX microparticles: the particle encapsulation. For example, micro/nano-particles
t50 for both SFN and celecoxib was <15 min at pH 2.5. can be encapsulated into the polymer matrix, which is
On the other hand, the t50 for HPMCAS microcapsules beneficial for drug delivery applications, especially in the
was ca. 20 min at pH 5.0 for both drugs. case of single emulsion. Since the particle matrix is either
The inner core of the W/O emulsion is composed of hydrophilic (W/O) or hydrophobic (O/W) in single emul-
hydrophilic materials, which is efficient for encapsulating sion, it is challenge to co-load hydrophilic and hydropho-
bioactive water-soluble agents, such as peptides, proteins, bic drugs.104 Therefore, the introduction of micro/nano-
antibodies and nucleic acids-based drugs, and even living particles into the polymer or lipid matrix compensates the
cells.100,101 The hydrophilic polymers, e.g., alginate (Fig. 4c),86 loading of drugs that do not dissolve in the inner phase.
chitosan102 and polyIJN-isopropylacrylamide) (PNIPA),103 were For example, mesoporous silicon (PSi) particles loaded
dissolved in water phase which subsequently broke into with hydrophilic drugs were dispersed into organic phase,
water droplets in the organic continuous phase by micro- enhancing the loading of hydrophilic drugs, such as ranit-
fluidics. After droplets formation, external triggers, such as idine and methotrexate, into a hydrophobic solid lipid
ionic environment and ultraviolet (UV) light, were with O/W emulsion.105 In addition, the encapsulation of
employed to induce the crosslinking of microgels.101 By PSi particles also shielded their free accessible pores and
simply adjusting the flow rates of the dispersed and con- controlled the drug release from PSi particles.83,106 Our
tinuous phases, and the gelation conditions, the morphol- group has encapsulated PSi nanoparticles in HPMCAS
ogy of sodium alginate (1.5 wt%) microgels was tuned polymer matrix, achieving the simultaneously loading and
from sphere to hemi-spherical, mushroom-like, disk-like or releasing of a hydrophilic drug, fluorouracil, and a hydro-
red blood cell-like (Fig. 4d). The release equilibrium (t100) phobic drug, celecoxib (Fig. 5a).90 Due to the pH respon-
of model drug, iopamidol, was ca. 100 min for the spheri- sive properties of the HPMCAS polymer, both payloads
cal alginate microgels; whereas a much faster drug release only released at pH > 6.5 with the t50 < 1 h and t100 ≈
(t100 ≈ 50 min) was observed for the mushroom-like algi- 2 h (Fig. 5b). Araújo and Santos et al.24,107 have success-
nate microgels (Fig. 4e). Compared to the polymeric fully co-delivered glucagon-like peptide-1 and dipeptidyl
microparticles produced by O/W emulsion, the cross-linked peptidase 4 enzyme inhibitor for diabetes therapy by oral
polymers in microgels presented looser networks, which drug delivery. The glucagon-like peptide-1 was loaded into
cannot fully prevent the release of small drug molecules. the PLGA or PSi nanoparticles, whose surface was

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1863
View Article Online

Tutorial review Lab on a Chip


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 4 Droplet based single and double emulsions for microcarriers fabrication. (a) Schematic illustration of PLGA microparticles fabricated by O/
W microfluidic single emulsion. (b) Release profiles of bupivacaine from the PLGA microparticles as a function of preparation methods and particle
size. Reprinted with permission from ref. 85, copyright 2009 Wiley-VCH. (c) Schematic illustration of alginate microgels prepared by the W/O
microfluidic single emulsion. (d) The viscosities and interfacial tension of the gelation bath controlled the morphologies of alginate microgels. (e)
Release behavior of iopamidol as a function of the morphologies of drug loaded alginate microgel. Reprinted with permission from ref. 86,
copyright 2012 AIP Publishing. (f) Schematic illustration of biodegradable lipid microcapsules prepared by W/O/W microfluidic double emulsions
for co-delivery of DOX (inner aqueous core) and PTX (lipid shell). (g) The release of payloads was temperature triggered at 37 °C. Reprinted with
permission from ref. 87, copyright 2013 ACS Publications. (h) Microgel capsule prepared by O/W/O microfluidic double emulsions with limited
interpenetration of the core and shell phases. (i) The release of Rhodamine B isothiocyanate–dextran was triggered by reswelling of the capsule
shell. Reprinted with permission from ref. 89, copyright 2010 ACS Publications.

sequentially modified with chitosan and cell-penetrating Several microfluidic methodologies are currently available
peptide. The co-delivery of atorvastatin and celecoxib with for the synthesis of Janus particles. For example, a
a precisely controlled ratio has been achieved by encapsu- T-junction microfluidics has been employed to fabricate
lating the atorvastatin-loaded halloysite nanotubes91 or PSi single emulsions; Janus droplets formed by the UV-directed
microparticles83 in HPMCAS using single emulsion droplet phase separation in the downstream channel.109 Through
microfluidics. the bonding of two polydimethylsiloxane layers,
Micro/nano-particles can also be encapsulated in double polyIJvinylidene fluoride–trifluoroethylene) ferroelectric poly-
emulsions. Herranz-Blanco and Santos et al.92 have encapsu- mers and Fe3O4 ferromagnetic particles were separately en-
lated PSi microparticles into phospholipid vesicles to sustain capsulated in the upper and lower layer of a single drop-
the release of piroxicam. The t50 for piroxicam-loaded PSi let.110 By simultaneously flowing two liquid monomers
microparticles was ca. 15 min for both pH 7.4 and 6.0. After containing photoinitiator into sodium dodecyl sulfate aque-
encapsulating the piroxicam-loaded PSi microparticles into ous solution in a T-junction microfluidic device, the Janus
the phospholipid vesicles, the t50 of piroxicam increased to droplets formed; Janus particles were made by UV-induced
ca. 30 at pH 6.0 and ca. 60 min at pH 7.4. Moreover, an all- crosslinking of the freshly formed Janus droplets.111 Khan
in-one microfluidic platform for co-delivery of multiple drugs et al.94 fabricated Janus particles to co-deliver ketoprofen
and particles have been developed by Kong et al.93 (Fig. 5c). and sodium fluorescein (Fig. 5e). The effect of the
DOX, erlotinib and 17-allyl-aminogeldanamycin were either collecting tube diameter on the morphology and payload re-
loaded into the PSi nanoparticles or directly loaded into the lease profiles of Janus particles with similar composition
core or shell of the giant liposome. The release profile of was studied. The large Janus particles (240 μm) showed non-
DOX from the giant liposome was sustained to be similar to uniform distribution of the two phases (Fig. 5f). For small
the poorly water-soluble erlotinib (Fig. 5d). particles (144 μm), uniform structure without defects was
Janus particles have unique asymmetric structure or observed (Fig. 5g). The release rate of ketoprofen was al-
functions, and these features can also be used in drug de- ways faster than that of hydrophilic sodium fluorescein
livery applications.79 By carefully controlling liquid flow in (Fig. 5h). The slower release rate of sodium fluorescein
channels to form Janus droplets, microfluidics offers a could be ascribed to its initial low loading amount (1 wt%)
unique and powerful tool to fabricate Janus particles.108 as compared to the 10 wt% of ketoprofen.

1864 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 5 Complexed microcarriers fabricated by droplet microfluidics for drug delivery. (a) Schematic illustration of encapsulating PSi nanoparticles
into pH responsive microparticle matrix. (b) The pH-responsive release of the payloads at neutral pH condition. Reprinted with permission from
ref. 90, copyright 2014 Wiley-VCH. (c) Multiple drugs and nanomaterials were encapsulated into giant liposome to achieve multiple-functions. (d)
Release profiles of payloads from the giant liposomes. Reprinted with permission from ref. 93, copyright 2015 Wiley-VCH. (e) Schematic illustration
of the capillary microfluidic setup for preparing drug loaded Janus particles. (f and g) The 240 μm Janus particles (f) showed dents while the 144
μm one (g) showed uniform structure without defects. (h) Release profiles of the prepared 240 and 144 μm Janus particles. Reprinted with permis-
sion from ref. 94, copyright 2014 Elsevier B.V.

3.4 Nanoparticles prepared by droplet microfluidics for the fusion chamber (Fig. 6f). After droplets merging, di-
controlled drug delivery methylsulfoxide was extracted into the aqueous phase,
resulting in the immediate formation of PLGA nano-
If the dispersed phase was partially miscible with the contin- particles.96 Because of the small volume of the droplets with
uous one, such as ethyl acetate (8.3 g in 100 mL) and di- the rapid mixing inside, the solvent extraction process oc-
methyl carbonate (13.9 g in 100 mL), the formed micro- curred immediately after droplets merging. Each merged
droplets containing particle precursors began to shrink after droplet acted as a microreactor in which numerous nano-
the initial creation of an interface. Theoretically, nano- particles formed; therefore, the production efficiency of
particles can be prepared by droplet microfluidics if low con- nanoparticles was supposed to be higher than the simple
centrations (e.g., 0.0002%, w/w)95 of particle precursors have droplet shrinking approach. With the help of extraction and
been employed. Alginate microgels were prepared by a single- evaporation approaches, particles of sizes ranging from 30
step droplet microfluidic technique, in which water and di- μm to 70 nm prepared (Fig. 6g). The obtained particles
methyl carbonate served as the dispersed and continuous showed narrow size distribution with less than 2% variation
phases, respectively (Fig. 6a). The on-chip cross-linking was in particle size.96 The complete release (t100) of fluorescein
performed by directly introducing CaCl2 into the alginate from PLGA nanoparticles (70 nm) with 85 : 15 and 50 : 50
droplets. Toward the preparation of alginate nanogels, the lactide to glycolide monomer ratios both took ca. 24 h
aqueous droplets containing low concentration of alginate (Fig. 6h). The t50 was ca. 4 and 3 h for the PLGA nano-
formed, shrank, and finally cross-linked by CaCl2 in the col- particles with 85 : 15 and 50 : 50 lactide to glycolide monomer
lection medium (Fig. 6b). Using droplet microfluidics, Ron- ratios, respectively.
deau et al.95 fabricated alginate microgels with the average In addition to the O/W droplet microfluidics, the use of
size ranging from 1 to 50 μm (Fig. 6c) and nanogels of sizes water in air (W/A) or organic solvent in air (O/A) droplets
ranging from 10 to 300 nm (Fig. 6d). Since the droplets were to prepare nanoparticles was proposed by Weitz's group.97
formed one by one with extremely low concentrations of algi- A microfluidic polydimethylsiloxane nebulator was fabri-
nate, the production efficiency of alginate nanogels by drop- cated using soft lithography to produce nanoparticles from
let microfluidics was usually limited and much lower than a wide range of materials.97 In the fabricated nebulator,
that of micro-ones. the fluid was intersected by a pair of additional fluids to
Hung et al.96 successfully constructed PLGA micro/nano- facilitate the mixing of fluid streams, initiating the possible
particles by shearing partially water miscible dimethyl car- chemical reactions prior to drop formation (Fig. 7a). The
bonate with aqueous solution (Fig. 6e). The dimethyl carbon- nebulator had six pairs of air inlets and the last junction
ate rapidly diffused and evaporated away, forming solid PLGA was a small inlet opening into a channel with larger di-
microparticles. Toward nanoparticle synthesis, the sequen- mensions. Benefiting from the supersonic air flow and the
tially formed water and dimethylsulfoxide droplets merged in small diameter, the freshly formed drops dried in a

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1865
View Article Online

Tutorial review Lab on a Chip


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 6 Micro/nano-particles prepared by droplet microfluidics. (a and b) Diagrams of the processes employed in the preparation of alginate
microgels (a) and nanogels (b). (c and d) The representative transmission electron microscopy (TEM) images of obtained alginate microgels (c, ca.
50 μm) and nanogels (d, ca. 200 nm). Reprinted with permission from ref. 95, copyright 2008 ACS publications. (e) Schematic illustration of device
design for the solvent evaporation to prepared PLGA microparticles. (f) Close-up of the microfluidic design to prepare PLGA nanoparticles. (g)
PLGA particles of sizes ranging from 30 μm to 70 nm obtained with the extraction or evaporation approaches. (h) Release profiles of fluorescein
from PLGA nanoparticles with a glycolic-to-lactic ratio of 50 : 50 and 85 : 15. Reprinted with permission from ref. 96, copyright 2010 The Royal So-
ciety of Chemistry.

superfast manner before the formation of crystal nuclei. loaded nanocapsules prepared at the gas pressure of 110
Amorphous nanoparticles possess favorable physicochemical and 510 kPa kept their structure after in vitro release
features, such as higher aqueous solubility and faster disso- studies (Fig. 7i). The zero-order release of amoxicillin from
lution rate, in comparison to the corresponding bulk crys- the resultant nanocapsules was observed post burst re-
tals. The nebulator produced amorphous NaCl nano- lease. Within 12 h, ca. 87% and 65% payloads were re-
particles when their diameter was below 15 nm, as leased from the nanocapsules with an average size of 92
illustrated in high-resolution TME (HRTEM) image (Fig. 7b) and 510 nm, respectively.98 As expected, the larger the
and the corresponding X-ray diffraction spectrum (Fig. 7c). particle size, the slower the release rate of payloads, which
The fenofibrate nanoparticles with an average diameter of can be ascribed to the long diffusion pathway for the
14 nm were always amorphous, as indicated by the lack of loaded drug molecules.112
any lattice plane in the HRTEM image (Fig. 7d) and the ab- By harnessing the in-fiber Rayleigh–Plateau capillary in-
sence of any peaks in X-ray diffraction spectrum (Fig. 7e). stability, Kaufman et al.99 fabricated the structured spheres
The small diameter of the obtained amorphous nano- with narrow size distribution. At first, a macroscopic scale
particles limited the probability of crystal nucleation. No preform model containing the intended particle cores em-
crystal nucleation was detected for all the prepared amor- bedded in a supporting cladding was prepared (Fig. 8a). In
phous nanoparticles under ambient conditions and at room the following process, the preformed model was thermally
temperature for at least 7 months. drawn into an extended fiber to reduce the diameter of
In 2013, Gunduz et al.98 successfully prepared ethyl cel- cores to the required size. An example of the fiber cross-
lulose nanocapsules from microfluidic generated micro- section, comprising a chalcogenide glass core entrapped in
bubbles (Fig. 7f and g). Specifically, the ethyl cellulose a polymer cladding (polyethersulphone), is shown in
ethanol solution and amoxicillin fluid flow in two outward Fig. 8a. After in situ cooling, the particles were frozen and
channels. The N2 gas flowing in a channel bisects the two released if needed. The fibers with cores were used to pro-
liquid flows and continuously generated ethyl cellulose duce particles, spanning an extremely wide range of sizes
microbubbles. The subsequent broken-up of the bubble from 2 mm down to 20 nm (Fig. 8b). The instability of a
surface resulted in the formation of ethyl cellulose nano- multimaterial fiber induced by the thermal process resulted
capsules. The gas pressure effectively controlled the size in a well-ordered emulsion. The structure of the prepared
and size distribution of the obtained nanocapsule. With a particles included core/shell (Fig. 8c), Janus (Fig. 8d) and
fixed flow rate of 500 μL min−1, the size of nanocapsules multi-section (Fig. 8e). Moreover, producing fibers with a
decreased from 550 ± 90 to 60 ± 10 nm by increasing the high density of cores allowed for an unprecedented level of
pressure from 110 to 510 kPa (Fig. 7h). Depending on parallelization.99 Specifically, one hundred million nano-
their size, the encapsulation efficiency of amoxicillin in particles with a diameter of 50 nm can prepared in meters-
the obtained nanocapsules was 65–88%. The amoxicillin- long fiber with a 25% fill factor.

1866 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 7 Nanoparticles prepared by the microfluidic nebulator and microbubbles. (a) Close-up of a microfluidic nebulator. Liquids were injected
through blue inlets and air was introduced through white inlets. (b) HRTEM image of spray-dried fenofibrate nanoparticles. (c) X-ray diffraction
spectra of fenofibrate crystals (3) and nanoparticles spray-dried with inlet air pressures of (1) 0.28 MPa and (2) 0.14 MPa. (d) HRTEM images of NaCl
nanoparticles spray-dried from aqueous solutions initially containing 4 mM NaCl. (e) X-ray diffraction spectra of (1) NaCl crystals, and spray-dried
NaCl nanoparticles produced from a solution containing (2) 40 mM and (3) 4 mM NaCl. Reprinted with permission from ref. 97, copyright 2015
AAAS. (f) V-junction microfluidic device to generate microbubbles. (g) Photographs showing the formation of microbubbles at t = 4 s. (h) SEM im-
ages of amoxicillin-loaded nanocapsules prepared at the gas pressure of 110 (upper, 510 nm) and 510 (lower, 90 nm) kPa after release studies, and
the corresponding size distribution histograms. (i) Amoxicillin release profiles from nanocapsule prepared at the gas pressure of 110 and 510 kPa.
Reprinted with permission from ref. 98, copyright 2013 Springer.

4 Precipitation and microfluidic ganic solvent to aqueous). In this case, the mixing time di-
hydrodynamic flow focusing rectly affects the final average size and size distribution of
the nanoparticles.113,115,116,118 In conventional bulk mixing
4.1 Mechanisms of microfluidic precipitation (conventional batch) methods, most nanoparticles are syn-
In the development of polymeric nanoparticles, one central thesized by altering the precursors into desired nano-
challenge is the precise engineering of nanoparticles with de- particles through stirring, shaking or sonicating. The gradu-
sired physicochemical characteristics in a reproducible ally changed ratio between the solvent and the anti-solvent
manner.113–116 The precursors can assemble into nano- forms a heterogeneous environment, leading to the forma-
particles when they experience a change in solvent quality. tion of polydispersed particles. Furthermore, the bulk
The self-assembly of block co-polymer into nanoparticles is mixing approach typically lacks precise control over the
currently believed to occur in three stages: (I) nucleation, (II) mixing process, which compromises the characteristics of
growth and (III) nanoparticle formation (Fig. 9a).52 First, sev- the obtained particles.113 In bulk synthesis of nanoparticles,
eral unimers of diblock polymers form a nucleus during a slow mixing implies that the aggregation of polymers to
solvent change. By adding more unimers, the size of the nu- form nanoparticles primarily occurs when mixing is incom-
clei continually grow through a diffusion-limited process in plete (Fig. 9b). Under this incomplete mixing condition,
the second stage. This growth process lasts until a polymer polymers easily adsorb on the nanoparticle aggregates, lead-
brush layer forms on the surface of a nanoparticle; and con- ing to formation of larger nanoparticles. In microfluidic de-
sequently, the addition of more unimers becomes difficult. vices, the mixing timescale can be tuned to be faster than
The third stage is featured by the extremely slow changes of the characteristic timescale for the nanoparticle precursors
nanoparticle sizes, which can be attributed to the equilib- to nucleate and grow. Therefore, the precipitation of nano-
rium of unimers exchange.117 particle precursors can occur primarily after the complete
An efficient way to accomplish the change in solvent solvent change with a lower fraction of organic solvent in
quality is mixing the solvent with anti-solvent (e.g., from or- the system. After complete mixing, polymers cannot easily

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1867
View Article Online

Tutorial review Lab on a Chip


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 8 Structured spheres generated by an in-fiber fluid instability. (a)


A macroscopic preform was thermally drawn into a fiber. The optical
micrograph of a fiber cross-section showed the core with a diameter
of ca. 20 mm. (b) Scanning electron microscopy (SEM) images of pre-
pared particles. (c) Front SEM views of two differently sized core/shell
particles. (d and e) Energy-dispersive X-ray diffraction (EDX) spectral
images (for sulphur, S) of an exposed Janus particle (d) and an exposed
‘beach ball’ particle (e) cross-section. Reprinted with permission from
ref. 99, copyright 2012 Nature Publishing Group.

be adsorbed or inserted into the nanoparticles, forming


more nanoparticles with smaller size (Fig. 9b).52
Because of the fast and efficient mixing, the physico- Fig. 9 Precipitation of polymers and the role of mixing time. (a) The
chemical features of the nanoparticles can be controlled by self-assembly of diblock co-polymer into nanoparticles. (b) Impact of
tuning the flow rate ratios between the non-solvent and sol- slow and rapid mixing on the nanoparticle preparation. Reprinted with
permission from ref. 52, copyright 2008 ACS publications.
vent or by changing the mixing patterns. For example, the
homogenous solvent environment enabled by the micro-
fluidic device for the nanoparticle precursors precipitation
yields smaller nanoparticles with narrower size distribution tions 4.3 and 4.4. A complete focusing of the nanoparticle
than those prepared by conventional methods.119,120 More- precursor solution into a small fluid filament can be achieved
over, microfluidic devices are usually operated in continuous by 3D-HFF, an ideal flow focusing approach. The 3D-HFF is
flow, which supports the high throughput production of more complicated to achieve than that of 2D-HFF. In the 3D-
nanoparticles with the same quality over time. These favor- HFF, also called coaxial microfluidic device, the central flow
able features on the production of polymeric nanoparticles is compressed both horizontally and vertically. Our group has
are important for the pharmaceutical industry.114 employed a microfluidic co-flow capillary device, constituted
of a tapered glass capillary coaxial aligned in another bigger
cylindrical one to achieve 3D-HFF (Fig. 10d).124,125 The fabri-
4.2 Overview of the HFF microfluidic systems for cated microcapillary device offers the distinct capability to
nanoparticle synthesis form truly 3D-coaxial flows, which is critical for the rapid and
Because of the advantages above-mentioned, microfluidic de- uniform mass transfer.126 In terms of polydimethylsiloxane
vices for the nanoparticle synthesis has been widely investi- devices, a sophisticated microfluidic approach to maintain
gated in past decades.113–116 One of the most common con- the 3D-focusing flow patterns have been utilized to prevent
tinuous flow mixing techniques in microfluidics is localized polymer aggregation near the microfluidic channel
hydrodynamic flow focusing (HFF). The HFF can be classified walls.126 We summarize the nanoparticles synthesized by 3D-
into two categories: two-dimensional (2D) and 3D HFF. Be- HFF microfluidic systems and the physicochemical properties
cause of the planar characteristics of most microfluidic de- of the obtained nanoparticles in Table 4 and discussed in de-
vices, 2D-HFF is usually focused in horizontal direction tail in section 4.5.
(Fig. 10a).121 To produce high-performance mixing, different
patterns for the 2D-HFF have been designed, such as the her-
ringbone mixer (Fig. 10b),70 and Tesla structured channel 4.3 Controlled drug delivery nanoparticles prepared by 2D-
(Fig. 10c).122,123 We summarize the nanoparticles synthesized HFF
by 2D-HFF microfluidic systems and the physicochemical Usually 2D-HFF is achieved by adding two additional inlets
properties, such as average particle size, size distribution (instead of one additional inlet) for the sheath liquid besides
(polydispersity, PDI) and release profiles, of the obtained the inlet for the nanoparticle precursor solution (Fig. 10a).
nanoparticles in Table 3 and discussed in detail in sec- The two sheath flows confine the nanoparticle precursor

1868 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

solution to the center of the channel in horizontal direction, 11 and 19 h for nanoparticles prepared by bulk and 2D-HFF,
resulting in a more uniform mixing time than that of respectively.
Y-junction devices.116 In addition, the confinement of the Because the combination of adequately selected drugs
nanoparticle precursors in horizontal direction by the sheath brings synergistic effects, there is an urgent need to develop
flows on the two sides, significantly reducing the mixing time robust approaches for co-delivery of therapeutics with differ-
by shortening the diffusion length.116 Flows in microfluidic ent physicochemical properties. To enhance the loading of
channels are laminar under typical operating conditions, water-soluble cisplatin in PLGA nanoparticles, its prodrug
thus the molecular diffusion across the fluids and the fluids platinumIJIV) [PtIJIV)] was conjugated to a polylactide (PLA) de-
homogenization is relatively slow for the 2D-HFF.70 The esti- rivative to form the PtIJIV) conjugated PLA (PLA–PtIJIV))
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

mated solvent mixing time was <0.4 ms (ref. 52) for 2D-HFF (Fig. 11e). A solution containing PLA–PtIJIV), PLGA-b-PEG and
by analogy to a previous study.117 Therefore, the mixing of docetaxel was converted to nanoparticles using a 2D-HFF
fluids was much faster than particle formation; consequently, microfluidic device. The high loading of both hydrophilic
the nanoparticles formed in a rapidly generated highly super- PtIJIV) and hydrophobic docetaxel was successfully achieved.
saturated solution aqueous of nanoparticle precursors. The obtained nanoparticles undergo controlled release of
A tunable mixing through 2D-HFF in microfluidic chan- both therapeutics over a period of 70 h (Fig. 11f).128
nels has been employed to control the precipitation of Chitosan is one of the most widely studied polysaccha-
PLGA15k-b-PEG4k diblock copolymers (Fig. 11a and b).52 By rides for biomedical applications. Dashtimoghadam et al.129
varying the flow rates, and the composition and concentra- synthesized a hydrophobic modified chitosan (HMCS) bio-
tion of the particle precursors, the obtained nanoparticles polymer, which can form nanoparticles in solvent free sys-
was optimized, showing smaller particle size and narrower tem. Using a classical 2D-HFF device, the HMCS formed
polydispersity than those prepared by conventional batch ap- nanoparticles by simply mixing the HMCS solution (pH = 5.5)
proach. Toward the PLGA–PEG nanoparticles prepared by 2D- with alkaline water (pH 9.0). The precipitation of HMCS was
HFF, the incorporation of PLGA increased the encapsulation driven by pH changes in a water environment, which was en-
efficiency of docetaxel from ca. 28% to 51% and the drug vironment friendly by avoiding the organic solvent.130 The
loading increased from ca. 4.1% to 6.8% (Fig. 11c). The incor- particle size, surface potential and compactness of the HMCS
poration of hydrophobic PLGA also prolonged the release of nanoparticles was controlled by the mixing time (2.5–75 ms)
docetaxel in the case of both bulk precipitation and 2D-HFF during HMCS precipitation.129,130 Regarding the formed par-
(Fig. 11d). The t50 of docetaxel was ca. 5–6 h for nanoparticles ticles, the higher the hydrophobicity of HMCS, the smaller
prepared by bulk and 2D-HFF without addition of PLGA. the particle size and the higher the compactness. The com-
Upon addition of PLGA, the t50 of docetaxel increased to ca. pactness of nanoparticles significantly affected their

Fig. 10 Overview of the HFF microfluidic systems toward the synthesis of nanoparticles. (a) Schematic diagram of Y-shaped and 2D-HFF micro-
fluidic setups. Reprinted with permission from ref. 116, copyright 2016 Elsevier B.V. (b) Schematic view of the herringbone mixer (channel with
ridges) and the three-dimensional twisting flow inside. Reprinted with permission from ref. 70, copyright 2002 AAAS. (c) Schematic illustration of a
2D-HFF microfluidic setup with Tesla structures, showing complete mixing at the fourth turn in the channel. Reprinted with permission from ref.
127, copyright 2010 ACS publications. (d) Schematic diagram of a 3D-HFF microfluidic device made up of capillaries. Reprinted with permission
from ref. 125, copyright 2015 Wiley-VCH.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1869
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Table 3 Summary of nanoparticles synthesized in 2D-HFF microfluidic platforms and the physicochemical properties of the obtained nanoparticles

Drug loading
Microfluidic devices Particle precursors degree Solvent/non-solvent Average size PDI Structure Estimated t50 and t100 Ref.
Tutorial review

Polydimethylsiloxane Polyij9,90-diIJ2- Perylene Tetrahydrofuran/water 30–80 nm 0.2 Sphere — 137


2D-HFF ethylhexyl)fluorene diimide dye,
ethynylene]/PEG —
Polydimethylsiloxane PLGA–PEG Docetaxel Acetonitrile/water 20–35 nm < that of bulk Sphere In PBS, t50 ≈ 5 h and ca. 52
2D-HFF 4.1% method 90% released in 72 h
Polydimethylsiloxane PLGA–PEG and PLGA Docetaxel, Acetonitrile/water 30–110 nm < that of bulk Sphere In PBS, t50 ≈ 20 h and 52
2D-HFF 6.8% method ca. 90% released in 72 h
Polydimethylsiloxane PLGA-b-PEG and PLA–PtIJIV) Docetaxel, 1% Acetonitrile/water ca. 100 nm 0.06–0.17 Sphere In PBS, t50 ≈ 12 h and 128

1870 | Lab Chip, 2017, 17, 1856–1883


2D-HFF t100 ≈ 72 h
Polydimethylsiloxane PLGA-b-PEG and PLA–PtIJIV) PtIJIV), 5% Acetonitrile/water ca. 100 nm 0.06–0.17 Sphere In PBS, t50 ≈ 40 h and 128
2D-HFF 90% released in 80 h
Polydimethylsiloxane Conjugation of PLGA, gold SFN, ca. Dimethylformamide and 85 nm 0.103 Sphere containing In PBS, t50 ≈ 3 h and 138
2D-HFF nanoparticles and 1.35% acetonitrile/lipid (40 μg mL−1) in gold t100 ≈ 500 h
hydroxypropyl β-cyclodextrin 30% methanol nanoparticles
Polydimethylsiloxane Conjugation of PLGA, gold DOX, ca. Dimethylformamide and 85 nm 0.103 Sphere containing In PBS, t50 ≈ 18 h and 138
2D-HFF nanoparticles and 0.52% acetonitrile/lipid (40 μg mL−1) in gold t100 < 150 h
hydroxypropyl β-cyclodextrin 30% methanol nanoparticles
Polydimethylsiloxane HMCS PTX, ca. 5.9% 1% acetic acid/basic water, flow 122 nm <0.16 Sphere In PBS, t50 ≈ 22 h and 129
2D-HFF ratio of 0.2 t100 ≈ 390 h
Polydimethylsiloxane HMCS PTX, ca. 8.2% 1% acetic acid/basic water, flow 96 nm <0.16 Sphere In PBS, t50 ≈ 55 h and 129
2D-HFF ratio of 0.075 ca. 70% released in 390
h
Polydimethylsiloxane HMCS PTX, ca. 8.7% 1% acetic acid/basic water, flow 74 nm <0.16 Sphere In PBS, t50 ≈ 22 h and 129
2D-HFF ratio of 0.031 ca. 85% released in 390
h
Polydimethylsiloxane Dendritic polyethylene and PTX, — Tetrahydrofuran/water, mixing 60–200 nm Close to 0.1 Sphere In PBS, t50 ≈ 35 h and 134
2D-HFF Poloxamer 407 time 2.5 ms ca. 80% released in 14
day
Polydimethylsiloxane Dendritic polyethylene and PTX, — Tetrahydrofuran/water, mixing 60–200 nm Close to 0.1 Sphere In PBS, t50 ≈ 58 h and 134
2D-HFF Poloxamer 407 time 13.6 ms ca. 70% released in 14
day
Polydimethylsiloxane Dendritic polyethylene and PTX, — Tetrahydrofuran/water, mixing 60–200 nm Close to 0.1 Core/shell sphere At pH 7.4, t50 ≈ 30 h 134
2D-HFF Poloxamer 407 time 76.0 ms and t100 ≈ 7 day
Polydimethylsiloxane PLGA-BP, SPION PTX, ca. 8% Tetrahydrofuran/water, flow ratio ca. 70 nm <0.13 SPION At pH 7.4, t50 ≈ 40 h 139
2D-HFF of 0.03 encapsulated and ca. 80% released in
sphere 120 h
Polydimethylsiloxane PLGA-BP, SPION PTX, ca. 7.5% Tetrahydrofuran/water, flow ratio ca. 75 nm <0.13 SPION At pH 7.4, t50 ≈ 60 h 139
2D-HFF of 0.07 encapsulated and ca. 75% released in
sphere 120 h
Polydimethylsiloxane PLGA-BP, SPION PTX, ca. 7.3% Tetrahydrofuran/water, flow ratio ca. 80 nm <0.13 SPION At pH 7.4, t50 ≈ 30 h 139
2D-HFF of 0.1 encapsulated and ca. 80% released in
sphere 120 h
Polydimethylsiloxane PLGA-BP, SPION PTX, ca. 5.8% Tetrahydrofuran/water, flow ratio ca. 105 nm <0.13 SPION At pH 7.4, t50 ≈ 20 h 139
2D-HFF of 0.2 encapsulated and ca. 85% released in
sphere 120 h
Polydimethylsiloxane PEO-b-PS functionalized Rhodamine B, Tetrahydrofuran/water 0.3–2.2 μm — Gold nanorods Linear increase of 140
2D-HFF gold nanorods and free — encapsulated payloads as a function of
PEO-b-PS vehicles NIR irradiation time
View Article Online

This journal is © The Royal Society of Chemistry 2017


Lab on a Chip
View Article Online

Lab on a Chip Tutorial review

Ref.
141

142

143
physicochemical features, such as drug release profiles and
stability. In comparison to the conventional approach, the en-

nanoparticles released in
payloads as a function of
Estimated t50 and t100 capsulation efficiency of PTX in HMCS nanoparticles pre-

NIR irradiation time


Linear increase of pared by the 2D-HFF device was significantly enhanced, pro-
viding a sustainable release profile with high tunability.129,131

<15% of gold
After the uptake of HMCS nanoparticles in tumor cells, the
encountered acidic endolysosomal environment triggered the
7 days release of PTX.130 The therapeutic efficiency of PTX was sig-
— nificantly enhanced after encapsulation in HMCS nano-
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

particles. Specifically, the half-maximal inhibitory concentra-


Janus-like vesicles

embedded sphere
Core/shell sphere

tion of PTX for MCF-7 breast cancer cells decreased from ca.
Gold nanorods

nanoparticles
encapsulated

1.0 to <0.001 nM post encapsulation.


Structure

In addition to PLGA and HMCS nanoparticles, poloxamer


407, hyaluronic acid and alginate nanoparticles have also
Gold

been successfully prepared by the 2D-HFF. The poloxamer


407 micelles prepared by the 2D-HFF showed higher control-
<Crosslinking

lability, better reproducibility, smaller size, and narrower


polydispersity than those prepared by the conventional ap-
method

proach.132 After microfluidic assembly to poloxamer 407 mi-


PDI

celles, mithramycin (a DNA-binding drug) presented a


slightly lower cytotoxicity and more pronounced differenti-
40–220 nm of
size and 0–65
Average size

ated activity compared to the free drugs.133 A nanocapsule


40–500 nm
1.86 ± 0.47
and 0.92 ±

nm shell
0.68 μm

consisting of a dendritic polyethylene core and a poloxamer


407 shell was synthesized by the 2D-HFF approach.134 The re-
sultant nanocapsule encapsulated large amounts of PTX (up
to ca. 20%, w/w), while providing a sustained release profile
pH 5.5 solution/pH 9.0 solution;

with high tunability.


6.5/Eudragit FS 30D solution

The crosslinked hyaluronic acid nanoparticles have also


PBS pH 7.4/spermine and

HMCS nanoparticles pH

been prepared by the 2D-HFF microfluidic device to encapsu-


Rhodamine B, Tetrahydrofuran/water
Solvent/non-solvent

late a magnetic resonance imaging contrast agent, gadolin-


PEG-PLL solution

ium diethylenetriamine penta-acetic acid (Gd–DTPA).135 The


adverse effects of Gd–DTPA on the nanoparticle preparation
was addressed by adjusting the process parameters, pH con-
ditions, and the hydrophilic/lipophilic balance of the surfac-
tants.132 The obtained nanoparticles presented sizes ranging
from 140 to 460 nm, depending on the operational condi-
nanoparticles
Drug loading

per mg PCPP

tions; low polydispersity, and negative zeta-potential,


5 mg gold

irrespectively of experimental conditions.135 Alginate is a bio-


PTX, —
degree

polymer with a favorable pH-responsive feature for oral deliv-


ery of peptides and proteins. Alginate nanogels have also


been synthesized by 2D-HFF approach with adjustable pore
HMCS and Eudragit FS 30D

size. The release of payloads from alginate nanogels were eas-


PEO-b-PS functionalized

Gold nanoparticles and


gold nanorods and free

ily tailored by tuning the pore size, which can be controlled


by adjusting the flow ratios and mixing time.136 The size of
Particle precursors

the obtained nanogels was in the range of 68–138 nm at flow


ratios of 0.02–0.2.136 The higher the flow ratio between the al-
PEO-b-PS

ginate fluid and sheath fluid, the smaller the size of the
nanogels and their compactness.
PCPP

By encapsulating nanoparticles into nanomatrices, the


obtained nano-in-nano composites may combine the favor-
Polydimethylsiloxane

Polydimethylsiloxane
Microfluidic devices
Table 3 (continued)

able features of both nanomaterials. The encapsulated nano-


herringbone mixer

2D-HFF with tesla

particles by microfluidics include gold,138,144 super-


Y-shaped with

paramagnetic iron oxide (SPIONs),139 quantum dots,144 PSi


(ref. 120) and drug nanoparticles.124,145 For example, a 2D-
2D-HFF

mixer

HFF approach was employed to co-deliver the SPIONs and


PTX in bisphosphonate (BP)-conjugated PLGA nanoparticles

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1871
View Article Online

Tutorial review Lab on a Chip

Table 4 Summary of polymeric nanoparticles synthesized in 3D-HFF microfluidic platforms and the physicochemical properties of the fabricated
nanoparticles

Particle Drug and Average Estimated t50


Microfluidic devices precursors loading degree Solvent/non-solvent size PDI Structure and t100 Ref.
Glass capillaries ADS-FA SFN, ca. 58.4% Acetone/water; 50% (v/v) ca. 233 <0.15 Core/shell At pH 5.0, t50 124
ethanol/water nm sphere ≈ 3 h and t100
≈8h
Glass capillaries HPMCAS SFN, ca. 45.2% Acetone/water (pH 10.5); 70–550 <0.2 Core/shell At pH 7.4, t50 145
water (pH 10.5)/water (pH nm sphere ≈ 9 min and
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

3.0) t100 ≈ 90 min


Glass capillaries HPMCAS PTX, ca. 42.6% Acetone/water (pH 10.5); 60–450 <0.2 Core/shell At pH 7.4, t50 145
water (pH 10.5)/water (pH nm sphere ≈ 4 min and
3.0) t100 ≈ 30 min
Glass capillaries AcDX and SFN, ca. 5.0% Ethanol/water ca. 350 <0.1 PSi At pH 5.0, t50 120
PSi nm, encapsulated ≈ 3.5 h and
nanoparticles sphere t100 ≈ 16 h
Glass capillaries AcDX and PTX, ca. 5.0% Ethanol/water ca. 350 <0.1 PSi At pH 5.0, t50 120
PSi nm, encapsulated ≈ 2.5 h and
nanoparticles sphere t100 ≈ 24 h
Glass capillaries AcDX and Methotrexate, Ethanol/water ca. 350 <0.1 PSi At pH 5.0, t50 120
PSi ca. 4.5% nm, encapsulated ≈ 2 h and t100
nanoparticles sphere ≈8h
Glass capillaries PHIS-b-PEG SFN, ca. 4.9% Ethanol and 0.1 M ca. 260 <0.2 PSi At pH 6.8, and 125
and hydrochloric acid/F127 nm encapsulated 5.5, t50 < 10
PLA-b-PEG solution (2 mg mL−1, pH nonspherical min and t100 <
12.8) nanoparticles 1h
Glass capillaries PLGA PTX, ca. 6% Acetonitrile/phospholipid < that Close to Sphere — 119
(50 μg mL−1) in 4% of bulk 0.1
ethanol method
Glass capillaries HMCS PTX, ca. 4% Acetonitrile and 1% (v/v) < that Close to Sphere — 119
acetic acid/phospholipid of bulk 0.1
in 4% ethanol, pH 9 method
Glass capillaries PLGA PTX, ca. 6% Acetonitrile/phospholipid < that Close to Sphere — 119
(50 μg mL−1) in 4% of bulk 0.1
ethanol, pH 8 method
Polydimethylsiloxane PLGA-b-PEG — Acetonitrile/water 30–230 < that of Sphere — 126
device nm 2D-HFF or
bulk
method
Polydimethylsiloxane PLGA-b-PEG Docetaxel, — Acetonitrile/water 20 and < that of Sphere At pH 7.4, t50 152
of 8 parallel 3D-HFF 35 nm 2D-HFF or ≈ 8 h and t100
bulk ≈ 24 h
method
Coaxial turbulent jet PLGA-b-PEG Docetaxel, ca. Acetonitrile/water <100 — Sphere — 154
mixer 1.7% nm
Coaxial turbulent jet PLGA-b-PEG Insulin, ca. 1.6% Dimethylsulfoxide/water <100 — Sphere — 154
mixer nm
Stainless steel PLGA Dexamethasone, Acetonitrile/water 35–350 — Sphere At pH 7.4, t50 159
capillaries <2% nm ≈ 10 min and
t100 ≈ 30 min

(SPION-PTX@PLGA-BP).139 The SPION-PTX@PLGA-BP polymeric core and SFN in the lipid corona (Fig. 12a). The
obtained in microfluidic rapid mixing regimen presented sev- nanocomposite core contained gold nanoparticles for imag-
eral favorable features, such smaller particle size, improved ing purposes and cyclodextrin molecules to maximize the
compactness of the nanoparticles, narrower size distribution DOX encapsulation. A fast release of the corona incorporated
and higher drug loading, over those prepared by the conven- SFN (t50 ≈ 3 h) and a delayed release of the core encapsu-
tional method. Toward biomedical applications, the SPION- lated DOX (t50 ≈ 18 h) were observed (Fig. 12b). The
PTX@PLGA-BP also showed a strong affinity to hydroxyapa- PLGA@Lipid nanocomposites were visualized with computed
tite, a prolonged circulation time in blood, high tumor locali- tomography (CT) due to the encapsulated gold nanoparticles.
zation and efficient suppression of bone metastatic tumor.139 In addition, the incorporation of Cy7-dye in the lipid corona
Besides SPIONs, gold nanoparticles were embedded into enabled the strong fluorescence of the nanocomposites in
polymeric nanomaterials by 2D-HFF. Mieszawska et al.138 near infrared (NIR) region, showing the accumulation at the
used chemically modified PLGA to formulate PLGA@Lipid tumor site by intravenously administration of the nano-
hybrid nanocomposites, containing doxorubicin (DOX) in the composites (Fig. 12c).138 2D-HFF was also used to precisely

1872 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 11 PLGA nanoparticles prepared by 2D-HFF. (a and b) A 2D-HFF microfluidic device (a) for preparation of PLGA–PEG nanoparticles (b). (c and
d) Encapsulation efficiency (c), drug loading (c) and release profiles (d) of PLGA–PEG nanoparticles. Reprinted with permission from ref. 52, copy-
right 2008 ACS publications. (e) Design and construction of PLGA–PEG nanoparticles co-loaded with PtIJIV) and docetaxel. (f) In vitro release profiles
of encapsulated PtIJIV) (circle) and docetaxel (square) from PLGA–PEG nanoparticles. Reprinted with permission from ref. 128, copyright 2010 United
States National Academy of Sciences.

control the assembly of block copolymer (PEO45-b-PS211) teth- of water fixed at 60 μL min−1. When the water flow rate in-
ered amphiphilic gold nanorods into various structures, such creased to 80 μL min−1, a similar trend on the change of vesi-
as spherical micelles, giant vesicles and raft-like disks cle size as a function of tetrahydrofuran flow rates was also
(Fig. 12d).140 Both the dimension and morphology of the detected. The hybrid Janus-like vesicles uniquely combined
nanoparticle self-assemblies were guided by simply varying the capability of autonomous propulsion, controlled by the
the flow rates of the fluids without templates. The balance concentration of the chemical fuel, and stimuli-responsive re-
between two competitive processes, the mixing of solvents lease of the encapsulated payloads, which can be externally
and the assembly kinetics of gold nanorods, was the most triggered by NIR light on demand.141 Upon the irradiation of
critical point to manipulate the self-assembly process. Gold NIR light, the intensity of Rhodamine B in the release me-
nanorods are featured by their strong photothermal effects in dium increased linearly as a function of irradiation time, and
the NIR range. Upon irradiation of NIR light, gold nanorods reached a plateau after about 70 min (Fig. 12i). The ability to
deformed to spherical gold nanoparticles (Fig. 12e), generat- use the flow to control the organization of nanoparticles into
ing extra spacing among gold nanoparticles and accelerating complex structures paves a new approach to fabricate struc-
the release of payloads. Without NIR laser irradiation, no ob- tured nanomaterials.
vious intensity of Rhodamine B was observed in the release Lipid−polymer and lipid−quantum dot (QD) nanoparticles
medium (Fig. 12f). However, the Rhodamine B concentration have been synthesized by a single mixing step of 2D-HFF.127
in water almost linearly increased with prolonged laser The obtained core/shell structured nanoparticle composed of
irradiation. a polymeric or a QD core, a hydrophilic polymeric shell, and
In addition to SPIONs and gold nanorods, a variety of in- a lipid monolayer at the interface of the core and the shell.
organic materials, including gold and platinum nano- The physicochemical properties of the obtained nanoparticles
particles, were introduced to the vesicle membrane of PEO45- were controlled by simply varying the composition and con-
b-PS455 (Fig. 12g).141 The introduction of metal nanoparticles centration of the nanoparticle precursors.127 Kim et al.144 pre-
to the vesicle membrane formed a distinct half of metal pared high-density lipoprotein (HDL)-mimicking nano-
nanoparticle-rich domain and imparted the vesicles with materials by 2D-HFF. Due to their antiatherothrombotic
multifunctionality. By increasing the concentration of PEO-b- properties, the reconstituted high-density lipoprotein (rHDL)
PS from 0.1 to 0.3 mg mL−1, the percentage of polymer-rich is considered as a natural treatment for cardiovascular dis-
domain coverage increased from ca. 15.4% to 40.4% eases. Different payloads of simvastatin (a hydrophobic
(Fig. 12h). Toward the obtained Janus-like vesicles, their aver- drug), gold nanoparticles, SPIONs, QDs or fluorophores were
age diameter can be precisely controlled by tuning the flow successfully entrapped into the μHDL nanoparticles. The
rates of fluids. After increasing the flow rate of tetrahydrofu- obtained nanomaterials showed the same properties, such as
ran from 20 to 50 μL min−1, the diameter of Janus-like vesi- size, morphology, and bioactivity, of the conventional rHDL
cles increased from ca. 749.2 to 2634.2 nm with the flow rate and the native HDL. This approach can contribute to the

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1873
View Article Online

Tutorial review Lab on a Chip

effective preparation of rHDL-based nanocomposites for (Fig. 13e). In the next step, the nanoemulsions were infused
medical applications.144 into a capillary microfluidic device, where droplets
A two-stage microfluidic device was developed to fabricate containing nanoemulsions were formed (Fig. 13f). With the
core/shell PLGA@Lipid hybrid nanoparticles (Fig. 13a).146 help of UV initiated free radical polymerization, the micro-
The low total flow rate of 41 mL h−1 resulted in the large droplets were hardened into Trojan particles (Fig. 13g). The
nanoparticles of 86.81 ± 1.59 nm in diameter with a PDI of polymerization process reduced the size of nanoemulsion
0.259 ± 0.025 (Fig. 13b and c). Nanoparticles synthesized at down to 20–32 nm for PEA and 10–15 nm for PMA. The Tro-
the flow rate of 246 mL h−1 were 62.5 ± 1.18 nm in diameter jan particles released the embedded nanoparticles upon con-
with a PDI of 0.173 ± 0.018, indicating a smaller particle size tact with the acidic release medium.148 At pH 6.8, it was
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

and narrower size distribution than that of nanoparticles syn- found that only 35% of encapsulated ketoprofen was released
thesized with a total flow rate of 41 mL h−1. All the synthe- over 24 h (Fig. 13h).
sized PLGA@Lipid showed core/shell structure (Fig. 13d). In
comparison to small PLGA@Lipid synthesized (62.5 ± 1.18
nm), the large ones (86.81 ± 1.59 nm) were more likely to ag- 4.4 Nanoparticles synthesized by fast mixing designs for 2D-
gregate in serum and exhibit a lower cellular uptake effi- HFF
cacy.146 Jiang's group has further developed this two-stage The herringbone mixer has attracted significant attention
microfluidic device to prepare nanoparticles with the same since its presentation from the Whitesides' group in 2002
chemical composition, size and surface properties, but vary- (Fig. 10b).70 The herringbone mixer is simple, elegant and
ing rigidity.147 It was found that the higher the rigidity of readily manufactured. For example, gold nanoparticles were
nanoparticles, the easier for nanoparticles to move through encapsulated into biodegradable polydiIJcarboxylatophenoxy)-
membranes and to be uptaken by the cells. Khan et al.148 phosphazene (PCPP) nanomatrix to form an inorganic–or-
prepared polyIJacrylamide) Trojan microparticles containing ganic hybrid nanocomposites (Au@PCPP).142 The size of
ketoprofen loaded polyIJethyl acrylate) (PEA) or polyIJmethyl Au@PCPP was controlled by the amount of polyIJethylene
acrylate) (PMA) nanoparticles. First, the polymerizable nano- glycol)-block-polylysine co-polymer (PEG-PLL). After inclusion
emulsions were produced in an elongational-flow micromixer of variable amounts of gold nanoparticles, the UV-vis

Fig. 12 HFF driven self-assembly of nanocomposites. (a) PLGA@Lipid nanocomposites composed of modified PLGA, lipids, gold nanoparticles,
DOX and SFN. (b) Release profiles of SFN and DOX from PLGA@Lipid nanocomposites in PBS at 37 °C. (c) The NIR fluorescence imaging of strong
Cy7 fluorescence indicated the accumulation of nanocomposites in tumor. Reprinted with permission from ref. 138, copyright 2013 ACS publica-
tions. (d) Self-assembly of amphiphilic gold nanorods tethered with block co-polymers, PEO45-b-PS211, under different 2D-HFF conditions. (e and
f) NIR-triggered the shape deformation of gold nanorods to spherical gold nanoparticles (e), generating extra spacing among gold nanoparticles
and accelerating the release of payloads (f). Reprinted with permission from ref. 140, copyright 2013 Wiley-VCH. (g) Self-assembly of a mixture of
building blocks into Janus-like vesicles by 2D-HFF. Upon the irradiation of NIR light, the self-propelled vesicles release payloads. (h) SEM images of
Janus-like vesicles containing 0.1 (top), 0.2 (middle) and 0.3 (bottom) mg mL−1 of PEO-b-PS. (i) The release profiles of Rhodamine B from Janus-
like vesicles as a function of time with laser on (square) and off (circle). Vesicles with full coverage of gold nanorods (triangle) with laser off served
as the control. Reprinted with permission from ref. 141, copyright 2013 Wiley-VCH.

1874 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

absorption peak of Au@PCPP was tuned into the NIR region. showed high cytocompatibility, while CPT-loaded ones
The amount of gold nanoparticles included also controlled significantly reduced the cell viability of prostate cancer cells.
the final size of the Au@PCPP nanocomposites.142 The gold A cancer targeting peptide (bombesin) was chemically at-
nanoparticle-encapsulated PCPP (Au@PCPP) nanocomposites tached to CPT-SPION@L-121 which can specifically bind and
produced strong CT contrast, serving as contrast agents. After be internalized by the prostate cancer cells. The CPT-
administration, Au@PCPP brook down into harmless by- SPION@L-121 generated significant T2-weighted magnetic
products and subsequently released the gold nanoparticles. resonance imaging contrast, allowing for direct monitoring
To prepare high-performance mixing, the 2D-HFF micro- of the biodistribution of the polymersomes.150 The self-
fluidic device has also been equipped with the Tesla struc- assembly of amphiphilic polyIJbutadiene)-block-PEO (PB-b-
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

tured channel (Fig. 10c).123 The proposed microfluidic device PEO) copolymer in water was studied using tetrahydrofuran
demonstrated excellent mixing performance after passing as co-solvent.151 Spherical micelles, vesicles and worm-like
only five mixing cell-pairs with Re ranging from 0.1 to 100. At micelles formed depending on the mixing principles, includ-
low flow rates, the microfluidic device showed a diffusion do- ing superfocus interdigital, slit interdigital, and caterpillar
main region, which moved to a convection domain region by micromixer.151 The self-assembly and in situ loading of PB-b-
increasing the flow rates. With contributions from both diffu- PEO polymersomes was performed in a continuous process
sion and convection, the mixing performance of 2D-HFF using a caterpillar micromixer. The membrane thickness and
equipped with a Tesla structured channel was similar to Tay- surface functionalities of the obtained polymersomes
lor dispersion.123 The high mixing efficiency of the proposed
2D-HFF equipped with a Tesla structured channel was further
proved by the recognition test between the epidermal growth
factor receptor L858R mutant specific rabbit monoclonal
antibody (a cancer biomarker) and the anti-rabbit IgG-
CFL555 and H1975 cells.122 The HMCS nanoparticles were
coated with a pH-sensitive copolymer (Eudragit FS 30D)
through a Tesla micromixer.143 Due to the acidic responsive
feature of HMCS, the oral administration of HMCS nano-
particles for targeted colorectal cancer therapy represents a
challenge. The enteric coating can protect the core HMCS
contents and prevent payload release in the severe pH condi-
tions of the stomach, while allowing the exposure of the core
content and release of drugs in colorectal section. The coat-
ing of HMCS nanoparticles with a pH-sensitive Eudragit layer
enabled the HMCS nanoparticles to bypass the acidic gastric
fluid without releasing the majority of the loaded PTX.143
A microfluidic origami device was designed to synthesize
DOX-loaded small PLGA (DOX@PLGA) nanoparticles through
enhanced mixing in microchannels.149 The studied geome-
tries of the microfluidic channels included flat, arc and dou-
ble spiral (Fig. 14a). For the flat microfluidic channel, the
particle size increased from ca. 100 to 234 nm by increasing
flow rate of organic solvent stream from 0.3 to 2.5 mL h−1
(Fig. 14b and c). At the flow rate of 2.5 mL h−1 for organic sol-
vent stream, the mixing time was 6 and 14.5 ms in 3D arc
and double spiral microchannels, respectively, which were
faster than that in 2D flat microchannels (54 ms). The Fig. 13 Two-stage microfluidic devices to prepare hybrid carriers. (a)
DOX@PLGA nanoparticles prepared by the 3D arc and double Schematic illustration of the two-stage microfluidic device for synthe-
spiral microchannels were around 100 nm (Fig. 14b and d) sizing the PLGA@Lipid hybrid nanoparticles. (b and c) Size distribution
(b) and polydispersity index (PDI; c) of PLGA@Lipid synthesized at dif-
with a narrow distribution (PDI < 0.06 for double spiral and
ferent flow rates. (d) TEM images of PLGA@Lipid synthesized at flow
PDI ≈ 0.13 for arc), exhibiting improved cellular uptake and rates of 41 and 246 mL h−1. Reprinted with permission from ref. 146,
anticancer efficacy. The production rate of one origami chip copyright 2015 AIP Publishing. (e and f) Microfluidic device toward the
was ca. 1.2 g per day at a maximum flow rate of 2.5 mL h−1 synthesis of nano-in-micro particles: (e) the elongation-flow micro-
with 2% PLGA and DOX solution.149 mixer for the production of nanoemulsions and (f) the single capillary
based droplet generator to prepare the nano-in-micro droplets. (g)
The caterpillar micromixer has also been used to prepare
SEM images of microparticles and their cross section at different mag-
polymersomes (Pluronic® L-121) simultaneously loaded with nifications exhibiting embedded nanoparticles. (h) The release profile
an anti-cancer drug (camptothecin, CPT) and SPIONs (CPT- of ketoprofen from Trojan microparticles in PBS of pH 6.8. Reprinted
SPION@L-121) (Fig. 14e).150 The drug-free polymersomes with permission from ref. 148, copyright 2015 Elsevier B.V.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1875
View Article Online

Tutorial review Lab on a Chip

depended on the length and end-groups of the block a production rate of 12.9 g h−1 with average diameters of 50
copolymer. and 85 nm.155
Most of the current microfluidic devices are made of
PMDS,128,130,144,156,157 however this material is sensitive to or-
4.5 Nanoparticles prepared by 3D-HFF for controlled drug ganic solvents, leading to swelling of microscale channels. Al-
delivery ternatively, glass shows chemically resistant features, excel-
To isolate the nanoparticle precursors from channel walls lent optical property and readily hydrophilic surface.23,38,158
and eliminate channel fouling, Rhee et al.126 developed a 3D- Our group has been employing glass capillaries to fabricate
HFF microfluidic device to synthesize PLGA-b-PEG nano- microfluidic devices. Moreover, our microfluidic devices offer
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

particles by nanoprecipitation. The isolation of the precipi- the distinct capability of creating truly 3D-coaxial flows,
tated nanoparticles in microfluidic channels was achieved by which are critical for the rapid and uniform mass transfer.126
the sequential inletting of bottom sheath, nanoparticle pre- Three types of nanoparticles, including PLGA, HMCS and
cursors and top sheath fluids. This 3D-HFF enabled robust AcDX, have been fabricated to demonstrate the versatility of
precipitation of nanoparticle precursors without polymer ag- this microfluidic platform.119 The PTX loading degree for all
gregation, independent of the polymer molecular weight or three nanoparticles was close to 6% (w/w). The production
precursor concentration.126 For example, PLGA-b-PEG nano- rate of one single microfluidic device reached up to 242.8 g
particles synthesized by 3D-HFF (30–230 nm) showed smaller per day. A model amphiphilic drug (dexamethasone) was also
particle size and narrower size distribution than that of 2D- incorporated into PLGA nanoparticles, using glass capillary
HFF (35–250 nm) or bulk synthesis (45–1130 nm). The parti- microfluidic devices.159
cle size of the nanoparticles prepared by the 3D-HFF was Our group has fabricated a nanocomposite consisting of
small enough for optimal uptake (i.e., <100 nm).126 an encapsulated PSi nanoparticle and an acid-degradable
The low production rate of nanoparticles limits the appli- AcDX matrix, by a 3D-HFF microfluidic glass capillary de-
cation of the microfluidics device for nanoparticle synthesis. vice.120 The obtained nano-in-nano composites, PSi@AcDX,
One strategy to enhance the production rate of nanoparticles showed enhanced surface smoothness, narrow size distribu-
is the parallelization of microfluidic devices using a multi- tion and considerably improved cytocompatibility. Three
layer system. With only 8 parallel 3D-HFF,152 PLGA-b-PEG therapeutics, PTX, SFN and methotrexate, with different phys-
nanoparticles were synthesized reproducibly with high pro- icochemical properties were simultaneously loaded into
duction rate up to 84 mg h−1. The parallelization of 3D-HFF PSi@AcDX with a ratiometric control. The release of all the
does not sacrifice the advantages of microfluidic technique, payloads was predominantly controlled by the decomposition
such as reproducibility, controllability and robustness.152 The of the outer AcDX matrix,120 regardless of the physico-
number of 3D-HFF parallel sets have been further amplified chemical properties of the drug molecules. In addition, a hy-
to 100 to prepare the methoxyl PLGA-b-PEG nanoparticles.153 brid nanocomposite comprised of a PSi nanoparticle and a
This increase in yield shortened the preparation time of the stimuli responsive polyIJL-histidine)-block-PEG (PHIS-b-PEG)
nanoparticles for a series of in vivo mice experiments (ca. 25 was also prepared by the 3D-HFF microfluidic device. Toward
mg) from over 5 h (ref. 126) to less than 20 min. The short the obtained PSi-based nanocomposite (PSi@PHIS-b-PEG),
preparation time is critical to minimize drug release during the formed micelles assembled on the surface of the PSi
nanoparticle synthesis. As a proof-of-concept, the pharmaco- nanoparticles. Benefiting from the pH-responsive feature of
kinetic and biodistribution study of small (20 and 35 nm) the micelles, the payloads were released upon exposure to
PLGA-b-PEG nanoparticles were performed on Balb/c mice.152 acidic conditions. The surface assembly of PHIS-b-PEG mi-
After intravenously injected to mice, both nanoparticles celles improved the surface smoothness of the PSi nano-
showed similar circulation profiles and PK parameters. particles, enhanced their stability in plasma, and improved
A versatile coaxial turbulent jet mixer154 was developed to their cytocompatibility.125 Recently, Fontana and Santos
synthesize a variety of nanoparticles, including PLGA–PEG, et al.160 prepared immunoadjuvant PSi-based nanovaccines
lipid vesicles, iron oxide and polystyrene nanoparticles, at by nanoprecipitation in a glass capillary microfluidic device.
high throughput up to 3 kg per day. The obtained nano- Vesicles, derived from cancer cell membranes, were utilized
particles showed high homogeneity, reproducibility and tun- to encapsulate the PSi or PSi@AcDX nanoparticles. The
ability, which were only accessible in specialized microscale obtained PSi-based nanocomposites were all biocompatible
mixing devices. Independently of the batch size, the high- over a wide range of concentrations. Most importantly, the
throughput formulated siRNA-polyelectrolyte polyplex nano- PSi-based nanocomposites present immunostimulant proper-
particles effectively inhibited the expression of luciferase ties, and consequently, they promoted the expression of co-
gene.154 Min et al.155 fabricated a pressure-tolerant 3D-paral- stimulatory signals and the secretion of pro-inflammatory cy-
lel polyimide film microreactor operating at up to 16 MPa tokines,160 important for cancer immunotherapy.
with direct 3D-HFF geometry for mass production of PLGA-b- Besides the PSi nanoparticles, our group has also fabri-
PEG nanoparticles. The multilayered polyimide film micro- cated a drug nanocrystal encapsulated core/shell nanovector
reactor fabricated by a simple one-step multilayer bonding by a two-step microfluidic nanoprecipitation (Fig. 15a).124
process, consisted of 8 sets of microchannels, which achieved The obtained nanovector consisted of a SFN nanocrystal core

1876 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 14 Microfluidic origami device and caterpillar micromixer. (a) Schematic illustration of microfluidic origami devices, including flat, arc and
double spiral geometries, for synthesizing PLGA nanoparticles. (b) Average size of PLGA nanoparticles as a function of flow rate in the origami chip
with different geometries. (c and d) SEM images of nanoparticles prepared in 2D flat (c) and 3D double spiral (d) channels at 2.5 mL h−1.
Reprinted with permission from ref. 149, copyright 2014 The Royal Society of Chemistry. (e) Continuous preparation of CPT-SPION@L-121 by a
caterpillar micromixer with twelve mixing steps. Reprinted with permission from ref. 150, copyright 2013 The Royal Society of Chemistry.

and a polymer (folic acid conjugated spermine-functionalized Recently, the superfast sequential nanoprecipitation
AcDX, ADS-FA) shell on a 1 : 1 ratio (HSFN@ADS-FA) method was developed to produce core/shell structured
(Fig. 15b). The favorable features of both polymer nano- nanocomposites.145 The microfluidic device consisted of
particles, such as biodegradability, stimuli-responsive release three sequentially nested cylindrical glass capillary tubes
of payloads, high stability in blood plasma, and ease of sur- (Fig. 15f). Clear core/shell structures for both PTX and
face functionalization, and drug nanocrystals, including SFN nanocrystals embedded HPMCAS nanoparticles
ultrahigh drug loading degree and fast dissolution, were (PTX@HPMCAS and SFN@HPMCAS) demonstrated the suc-
inherited by the prepared nano-in-nano vector HSFN@ADS- cessful encapsulation of drug nanocrystals (Fig. 15g).
FA. Consequently, a small amount of nanocarriers was Benefiting from the multiplexed microfluidic design, the
needed to deliver a clinically relevant therapeutic dose, reduc- time intervals between two sequential nanoprecipitation
ing the potential side effects of nanocarriers and treatment processes were extremely short, resulting in the superfast
cost. At both pH 7.4 and 5.0, SFN completely released from formation of the outer layer onto the surface of freshly the
the bare SFN nanocrystals within 0.5 h (Fig. 15c). In contrast, formed nanocomposite cores. The superfast deposition of
the HSFN@ADS-FA only released ≈9.3% of payloads after 24 the outer polymer layer avoided the instability issues of the
h at pH 7.4. After decreasing the pH value to 5.0, a complete nanocomposite's cores without using any stabilizers. Barely
release of payloads from the HSFN@ADS-FA was achieved no drug was released from the PTX@HPMCAS and
within only 2 h (Fig. 15d). The half-maximal inhibitory con- SFN@HPMCAS at pH 1.2 and 5.0 (Fig. 15h), indicating the
centration of the drug nanocrystal encapsulated nano-in- tightly entrapment of the drug nanocrystals inside the
nano vector was ca. 54 times lower than the conventional HPMCAS polymer shell. After increasing the pH to 7.4, the
nanovector with a low drug loading degree (Fig. 15e). These fast and complete release of the payloads was observed,
properties of HSFN@ADS-FA could significantly reduce the which was attributed to the dissolution of HPMCAS, and
side effects caused by the loaded drug molecules and en- consequently, the exposure of the encapsulated drug nano-
hance the therapeutic efficiency of payloads. crystals into the release medium.145

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1877
View Article Online

Tutorial review Lab on a Chip


Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Fig. 15 Fabrication of core/shell nanoparticles by multistep and superfast sequential microfluidic nanoprecipitation. (a and b) Schematic view of
the two-step microfluidic nanoprecipitation (a) to prepare SFN@ADS-FA nanovector (b). (c and d) The release profiles of SFN from SFN nano-
crystals (c) and SFN@ADS-FA (d) at pH 7.4 and 5.0. (e) The half-maximal inhibitory concentration (IC50) of the nanomaterials without FA pre-
incubation was compared with that after FA pre-incubation. Reprinted with permission from ref. 124, copyright 2017 Wiley-VCH. (f) Schematic view
of the 3D-glass capillary microfluidic device to implement the superfast sequential nanoprecipitation (not to scale). (g) TEM images of
PTX@HPMCAS (1) and SFN@HPMCAS (2) nanocomposites. (h) Drug release profiles of the core/shell nanocomposites with continuous changes in
the pH conditions from pH 1.2 to 7.4. Reprinted with permission from ref. 145, copyright 2017 ACS publications.

5 Discussion and perspective tion, structure, composition, surface properties and payload
release profiles of the obtained carriers all can be controlled
Microfluidic technologies have shown great potential in syn- by tuning microfluidic parameters.21,23 By assembling a vari-
thesizing micro/nano-carriers for controlled drug delivery ap- ety of synthetic procedures into a single microfluidic device,
plications.28 Toward droplet microfluidics, a wide variety of the one-step preparation of micro/nano-particles with com-
microcarriers, including microparticles, microcapsules and plex structures can be easily achieved.145 In comparison to
microgels, have been successfully fabricated.46 The obtained batch-type bulk synthesis methods, the continuous synthesis
microcarriers are featured with controlled size, structure, of carriers offered by microfluidics leads to high batch-to-
composition, surface properties and controlled release of pay- batch reproducibility.119,156 These favorable features make
loads. Due to the size limit of droplets, most of the carriers microfluidics an ideal platform for controlled synthesis of
prepared by droplet microfluidics are in micrometer scale micro/nano-particles for controlled drug delivery
(>1 μm). Nanoparticles can also be prepared by droplet applications.
microfluidics, when the concentration of particle precursors Notwithstanding the recent advances on the fabrication of
is low enough.95 The low concentration of particle precursors drug delivery systems by microfluidics, the translation of this
enables tens to hundreds of times of shrinking of the formed technology from academic research to the industrial and clin-
microdroplets to nanoscale. Because of the low concentra- ical practice still faces several critical challenges. One of the
tions of particle precursors, the production rate of nano- greatest challenges is the quantity of drug delivery systems
particles by droplet microfluidics is usually much lower than synthesized by microfluidic devices, whose daily production
that of micro-ones. HFF has been widely used to synthesize rate is usually in the milligram range.156 However, the desir-
nanoparticles.116 The successful synthesis of nanoparticles by able carrier production rate for clinical studies and
HFF can be attributed to the rapid mixing of solvent and industrial-scale production is in the order of kilograms per
non-solvent in microfluidic channels. Toward HFF, especially day.154 One of most promising approach to prepare an indus-
3D-HFF, the mixing time of solvent and non-solvent is faster trial scale production of carriers by microfluidics is the para-
than the nucleation time of nanoparticle precursors, llelization of microfluidic channels.161,162 The parallelization
resulting in the formation of small nanoparticles with narrow of microfluidic channels can directly multiply the daily pro-
size distribution. duction rate of carriers with the same properties as those pre-
Benefiting from the accurate manipulation of fluids in pared at the bench scale. Recently, a versatile coaxial turbu-
microchannels and consequently the fine modulation of syn- lent jet mixer has been developed to synthesize a variety of
thesis conditions, microfluidics enables the precise control nanoparticles with the production rate up to 3 kg per day,
over physicochemical properties of the obtained micro/nano- which is suitable for clinical trials and industrial-scale
particles.19,23,28 Specifically, the average size, size distribu- production.

1878 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

The fluid flow inside microchannels is usually con- 2 M. W. Tibbitt, J. E. Dahlman and R. Langer, J. Am. Chem.
trolled by pumps, such as syringe or peristaltic pumps. Soc., 2016, 138, 704–717.
Due to the small dimension of the microchannels, the 3 S. Mitragotri, D. G. Anderson, X. Chen, E. K. Chow, D. Ho,
pressure inside the microfluidic device rises dramatically A. V. Kabanov, J. M. Karp, K. Kataoka, C. A. Mirkin, S. H.
as a function of the increasing flow rate.119 The linear Petrosko, J. Shi, M. M. Stevens, S. Sun, S. Teoh, S. S.
force afforded by the fluid controlled apparatus and the Venkatraman, Y. Xia, S. Wang, Z. Gu and C. Xu, ACS Nano,
withstanding pressure of the microfluidic device together 2015, 9, 6644–6654.
determine the highest flow rates for a certain microfluidic 4 R. Tong and R. Langer, Cancer J., 2015, 21, 314–321.
system. Toward the high throughput production of drug 5 A. C. Anselmo and S. Mitragotri, AAPS J., 2015, 17,
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

delivery systems, the industrial-scale fluid control appara- 1041–1054.


tus should be employed. The carrier synthesis process by 6 O. Veiseh, B. C. Tang, K. A. Whitehead, D. G. Anderson and
microfluidics usually uses large amounts of water and or- R. Langer, Nat. Rev. Drug Discovery, 2015, 14, 45–57.
ganic solvents. Purification is required to remove the re- 7 G. G. Walmsley, A. McArdle, R. Tevlin, A. Momeni, D.
sidual organic solvents and unencapsulated drugs from Atashroo, M. S. Hu, A. H. Feroze, V. W. Wong, P. H. Lorenz,
the obtained formulations. The suitable purification M. T. Longaker and D. C. Wan, Nanomed. Nanotechnol. Biol.
methods to treat large volumes of formulation include ul- Med., 2015, 11, 1253–1263.
trafiltration,163 diafiltration164 and cross-flow micro- 8 R. N. H. Fang and L. F. Zhang, Annu. Rev. Chem. Biomol.
filtration.165,166 The purification of these obtained formula- Eng., 2016, 7, 305–326.
tions demands standard protocols for industrial 9 O. Veiseh, J. C. Doloff, M. Ma, A. J. Vegas, H. H. Tam, A. R.
applications. Considering the advantages and challenges Bader, J. Li, E. Langan, J. Wyckoff, W. S. Loo, S.
offered by microfluidics, it is quite evident that the Jhunjhunwala, A. Chiu, S. Siebert, K. Tang, J. Hollister-
microfluidic-assisted fabrication of drug delivery systems Lock, S. Aresta-Dasilva, M. Bochenek, J. Mendoza-Elias, Y.
has already an important impact on the field of controlled Wang, M. Qi, D. M. Lavin, M. Chen, N. Dholakia, R.
drug delivery. Thakrar, I. Lacik, G. C. Weir, J. Oberholzer, D. L. Greiner,
R. Langer and D. G. Anderson, Nat. Mater., 2015, 14,
6 Conclusions 643–651.
10 F. Fontana, D. Liu, J. T. Hirvonen and H. A. Santos, Wiley
We have reviewed the potential benefits brought by the Interdiscip. Rev.: Nanomed. Nanobiotechnol., 2017, 9, e1421.
microfluidic techniques toward the production of polymeric 11 B. Lee Chung, M. J. Toth, N. Kamaly, Y. J. Sei, J. Becraft,
drug delivery systems. The microfluidic-based systems open W. J. M. Mulder, Z. A. Fayad, O. C. Farokhzad, Y. T. Kim
new strategies regarding the fabrication of complex struc- and R. Langer, Nano Today, 2015, 10, 759–776.
tured carriers for controlled drug delivery compared to the 12 E. Blanco, H. Shen and M. Ferrari, Nat. Biotechnol.,
conventional batch methods. In parallel to the droplet micro- 2015, 33, 941–951.
fluidics for microparticles preparation, the synthesis of nano- 13 Z. H. Tang, C. L. He, H. Y. Tian, J. X. Ding, B. S. Hsiao, B.
particles in microfluidic devices is also becoming an attrac- Chu and X. S. Chen, Prog. Polym. Sci., 2016, 60, 86–128.
tive research area. The research in the past decade has 14 S. Verma, R. Gokhale and D. J. Burgess, Int. J. Pharm.,
demonstrated that microfluidics is a versatile approach to 2009, 380, 216–222.
precisely control the physicochemical properties of the pro- 15 B. K. Lee, Y. Yun and K. Park, Adv. Drug Delivery Rev.,
duced drug delivery carriers, even though the synthesis of 2016, 107, 176–191.
carriers is limited by the low production rate. Overall, the 16 H. K. Makadia and S. J. Siegel, Polymer, 2011, 3, 1377–1397.
microfluidic technology in controlled drug delivery applica- 17 E. Lepeltier, C. Bourgaux and P. Couvreur, Adv. Drug
tions brings exciting opportunities to expand the body of Delivery Rev., 2014, 71, 86–97.
knowledge in the field and advance to the clinical translation 18 R. J. Cohrs, T. Martin, P. Ghahramani, L. Bidaut, P. J. Higgins
of drug delivery systems. and A. Shahzad, New Horiz. Transl. Med., 2015, 2, 86–88.
19 Q. Feng, J. Sun and X. Jiang, Nanoscale, 2016, 8,
Acknowledgements 12430–12443.
We acknowledge financial support from the Jane and Aatos 20 A. Choi, K. D. Seo, D. W. Kim, B. C. Kim and D. S. Kim, Lab
Erkko Foundation (Grant No. 4704010 and 4704482), the Uni- Chip, 2017, 17, 591–613.
versity of Helsinki Research Funds, the Academy of Finland 21 W. Wang, M. J. Zhang and L. Y. Chu, Acc. Chem. Res.,
(Grant No. 304844 and 297580), and the European Research 2014, 47, 373–384.
Council (Grant No. 310892). 22 C. Berkland, M. King, A. Cox, K. Kim and D. W. Pack,
J. Controlled Release, 2002, 82, 137–147.
References 23 W. J. Duncanson, T. Lin, A. R. Abate, S. Seiffert, R. K. Shah
and D. A. Weitz, Lab Chip, 2012, 12, 2135–2145.
1 J. R. Heath, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, 24 F. Araújo, N. Shrestha, M. A. Shahbazi, D. Liu, B. Herranz-
14436–14443. Blanco, E. M. Mäkilä, J. J. Salonen, J. T. Hirvonen, P. L.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1879
View Article Online

Tutorial review Lab on a Chip

Granja, B. Sarmento and H. A. Santos, ACS Nano, 2015, 9, 51 J. D. Wang, W. W. Chen, J. S. Sun, C. Liu, Q. F. Yin, L.
8291–8302. Zhang, Y. L. Xianyu, X. H. Shi, G. Q. Hu and X. Y. Jiang,
25 M. A. Stuart, W. T. Huck, J. Genzer, M. Muller, C. Ober, M. Lab Chip, 2014, 14, 1673–1677.
Stamm, G. B. Sukhorukov, I. Szleifer, V. V. Tsukruk, M. 52 R. Karnik, F. Gu, P. Basto, C. Cannizzaro, L. Dean, W. Kyei-
Urban, F. Winnik, S. Zauscher, I. Luzinov and S. Minko, Manu, R. Langer and O. C. Farokhzad, Nano Lett., 2008, 8,
Nat. Mater., 2010, 9, 101–113. 2906–2912.
26 S. Mura, J. Nicolas and P. Couvreur, Nat. Mater., 2013, 12, 53 M. T. Zhu, G. J. Nie, H. Meng, T. Xia, A. Nel and Y. L. Zhao,
991–1003. Acc. Chem. Res., 2013, 46, 622–631.
27 G. M. Whitesides, Nature, 2006, 442, 368–373. 54 X. L. Huang, X. Teng, D. Chen, F. Q. Tang and J. Q. He,
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

28 F. Fontana, M. P. A. Ferreira, A. Correia, J. T. Hirvonen and Biomaterials, 2010, 31, 438–448.


H. A. Santos, J. Drug Delivery Sci. Technol., 2016, 34, 76–87. 55 Y. Z. He and K. Park, Mol. Pharmaceutics, 2016, 13,
29 T. M. Squires and S. R. Quake, Rev. Mod. Phys., 2005, 77, 2164–2171.
977–1026. 56 S. E. Gratton, P. A. Ropp, P. D. Pohlhaus, J. C. Luft, V. J.
30 E. K. Sackmann, A. L. Fulton and D. J. Beebe, Nature, Madden, M. E. Napier and J. M. DeSimone, Proc. Natl.
2014, 507, 181–189. Acad. Sci. U. S. A., 2008, 105, 11613–11618.
31 P. N. Nge, C. I. Rogers and A. T. Woolley, Chem. Rev., 57 K. C. L. Black, Y. C. Wang, H. P. Luehmann, X. Cai, W. X.
2013, 113, 2550–2583. Xing, B. Pang, Y. F. Zhao, C. S. Cutler, L. H. V. Wang, Y. J.
32 A. J. L. Morgan, L. H. San Jose, W. D. Jamieson, J. M. Liu and Y. N. Xia, ACS Nano, 2014, 8, 4385–4394.
Wymant, B. Song, P. Stephens, D. A. Barrow and O. K. 58 Y. Geng, P. Dalhaimer, S. S. Cai, R. Tsai, M. Tewari, T.
Castell, PLoS One, 2016, 11, e0152023. Minko and D. E. Discher, Nat. Nanotechnol., 2007, 2,
33 H. W. Li, Y. Q. Fan, R. Kodzius and I. G. Foulds, Microsyst. 249–255.
Technol., 2012, 18, 373–379. 59 S. I. Simon and G. W. Schmidschonbein, Biophys. J.,
34 S. Begolo, G. Colas, J. L. Viovy and L. Malaquin, Lab Chip, 1988, 53, 163–173.
2011, 11, 508–512. 60 C. B. He, Y. P. Hu, L. C. Yin, C. Tang and C. H. Yin,
35 D. Qin, Y. N. Xia and G. M. Whitesides, Nat. Protoc., Biomaterials, 2010, 31, 3657–3666.
2010, 5, 491–502. 61 R. A. Yokel, Nanomed. Nanotechnol. Biol. Med., 2016, 12,
36 J. C. McDonald and G. M. Whitesides, Acc. Chem. Res., 2081–2093.
2002, 35, 491–499. 62 M. Beck-Broichsitter, J. Nicolas and P. Couvreur, Eur. J.
37 T. Odera, H. Hirama, J. Kuroda, H. Moriguchi and T. Torii, Pharm. Biopharm., 2015, 97, 304–317.
Microfluid. Nanofluid., 2014, 17, 469–476. 63 D. H. Jo, J. H. Kim, T. G. Lee and J. H. Kim, Nanomed.
38 R. K. Shah, H. C. Shum, A. C. Rowat, D. Lee, J. J. Agresti, Nanotechnol. Biol. Med., 2015, 11, 1603–1611.
A. S. Utada, L. Y. Chu, J. W. Kim, A. Fernandez-Nieves, 64 H. Kettiger, A. Schipanski, P. Wick and J. Huwyler, Int. J.
C. J. Martinez and D. A. Weitz, Mater. Today, 2008, 11, Nanomed., 2013, 8, 3255–3269.
18–27. 65 L. Zhang, Q. Feng, J. L. Wang, J. S. Sun, X. H. Shi and X. Y.
39 C. Iliescu, H. Taylor, M. Avram, J. M. Miao and S. Franssila, Jiang, Angew. Chem., Int. Ed., 2015, 54, 3952–3956.
Biomicrofluidics, 2012, 6, 16505. 66 Q. Feng, J. P. Liu, X. Y. Li, Q. H. Chen, J. S. Sun, X. H. Shi,
40 B. Herranz-Blanco, E. Ginestar, H. B. Zhang, J. Hirvonen B. Q. Ding, H. J. Yu, Y. P. Li and X. Y. Jiang, Small,
and H. A. Santos, Int. J. Pharm., 2017, 516, 100–105. 2017, 13, 1603109.
41 C. X. Zhao, Adv. Drug Delivery Rev., 2013, 65, 1420–1446. 67 L. Zhang, Q. Feng, J. Wang, S. Zhang, B. Ding, Y. Wei, M.
42 I. U. Khan, C. A. Serra, N. Anton and T. Vandamme, Dong, J. Y. Ryu, T. Y. Yoon, X. Shi, J. Sun and X. Jiang, ACS
J. Controlled Release, 2013, 172, 1065–1074. Nano, 2015, 9, 9912–9921.
43 J. N. Lee, C. Park and G. M. Whitesides, Anal. Chem., 68 D. H. Liu, F. X. Liu, Z. H. Liu, L. L. Wang and N. Zhang,
2003, 75, 6544–6554. Mol. Pharmaceutics, 2011, 8, 2291–2301.
44 E. Sollier, C. Murray, P. Maoddi and D. Di Carlo, Lab Chip, 69 E. A. Sykes, J. Chen, G. Zheng and W. C. W. Chan, ACS
2011, 11, 3752–3765. Nano, 2014, 8, 5696–5706.
45 K. N. Ren, Y. Chen and H. K. Wu, Curr. Opin. Biotechnol., 70 A. D. Stroock, S. K. W. Dertinger, A. Ajdari, I. Mezic, H. A.
2014, 25, 78–85. Stone and G. M. Whitesides, Science, 2002, 295, 647–651.
46 P. Zhu and L. Wang, Lab Chip, 2016, 17, 34–75. 71 K. V. Sharp and R. J. Adrian, Exp. Fluids, 2004, 36,
47 S. Mazzitelli, L. Capretto, F. Quinci, R. Piva and C. 741–747.
Nastruzzi, Adv. Drug Delivery Rev., 2013, 65, 1533–1555. 72 J. Atencia and D. J. Beebe, Nature, 2005, 437, 648–655.
48 N. T. Nguyen, S. A. Shaegh, N. Kashaninejad and D. T. 73 J. K. Nunes, S. S. Tsai, J. Wan and H. A. Stone, J. Phys. D:
Phan, Adv. Drug Delivery Rev., 2013, 65, 1403–1419. Appl. Phys., 2013, 46, 114002.
49 H. A. Stone, A. D. Stroock and A. Ajdari, Annu. Rev. Fluid 74 A. S. Utada, A. Fernandez-Nieves, H. A. Stone and D. A.
Mech., 2004, 36, 381–411. Weitz, Phys. Rev. Lett., 2007, 99, 094502.
50 H. F. Chan, S. Y. Ma and K. W. Leong, Regener. Biomater., 75 P. Garstecki, M. J. Fuerstman, H. A. Stone and G. M.
2016, 3, 87–98. Whitesides, Lab Chip, 2006, 6, 437–446.

1880 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

76 S. H. Kim, J. W. Kim, J. C. Cho and D. A. Weitz, Lab Chip, 99 J. J. Kaufman, G. M. Tao, S. Shabahang, E. H. Banaei,
2011, 11, 3162–3166. D. S. S. Deng, X. D. Liang, S. G. Johnson, Y. Fink and A. F.
77 L. L. A. Adams, T. E. Kodger, S. H. Kim, H. C. Shum, T. Abouraddy, Nature, 2012, 487, 463–467.
Franke and D. A. Weitz, Soft Matter, 2012, 8, 100 S. Utech, R. Prodanovic, A. S. Mao, R. Ostafe, D. J. Mooney
10719–10724. and D. A. Weitz, Adv. Healthcare Mater., 2015, 4, 1628–1633.
78 C. H. Chen, R. K. Shah, A. R. Abate and D. A. Weitz, 101 E. Tumarkin and E. Kumacheva, Chem. Soc. Rev., 2009, 38,
Langmuir, 2009, 25, 4320–4323. 2161–2168.
79 S. Yang, F. Guo, B. Kiraly, X. Mao, M. Lu, K. W. Leong and 102 V. Zamora-Mora, D. Velasco, R. Hernandez, C. Mijangos
T. J. Huang, Lab Chip, 2012, 12, 2097–2102. and E. Kumacheva, Carbohydr. Polym., 2014, 111, 348–355.
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

80 T. Nisisako, T. Torii, T. Takahashi and Y. Takizawa, Adv. 103 D. Jagadeesan, I. Nasimova, I. Gourevich, S. Starodubtsev
Mater., 2006, 18, 1152–1156. and E. Kumacheva, Macromol. Biosci., 2011, 11, 889–896.
81 R. K. Shah, J. W. Kim and D. A. Weitz, Adv. Mater., 104 D. Liu, L. M. Bimbo, E. M. Mäkilä, F. Villanova, M.
2009, 21, 1949–1953. Kaasalainen, B. Herranz-Blanco, C. M. Caramella, V. P.
82 J. A. Champion, Y. K. Katare and S. Mitragotri, J. Controlled Lehto, J. Salonen, K. H. Herzig, J. T. Hirvonen and H. A.
Release, 2007, 121, 3–9. Santos, J. Controlled Release, 2013, 170, 268–278.
83 D. Liu, H. Zhang, B. Herranz-Blanco, E. M. Mäkilä, V. P. 105 D. Liu, B. Herranz-Blanco, E. M. Mäkilä, L. R. Arriaga, S.
Lehto, J. Salonen, J. T. Hirvonen and H. A. Santos, Small, Mirza, D. A. Weitz, N. Sandler, J. Salonen, J. T. Hirvonen
2014, 10, 2029–2038. and H. A. Santos, ACS Appl. Mater. Interfaces, 2013, 5,
84 R. Vasiliauskas, D. Liu, S. Cito, H. Zhang, M. A. Shahbazi, 12127–12134.
T. Sikanen, L. Mazutis and H. A. Santos, ACS Appl. Mater. 106 D. Liu, E. M. Mäkilä, H. Zhang, B. Herranz, M.
Interfaces, 2015, 7, 14822–14832. Kaasalainen, P. Kinnari, J. Salonen, J. T. Hirvonen and
85 Q. Xu, M. Hashimoto, T. T. Dang, T. Hoare, D. S. Kohane, H. A. Santos, Adv. Funct. Mater., 2013, 23, 1893–1902.
G. M. Whitesides, R. Langer and D. G. Anderson, Small, 107 F. Araújo, N. Shrestha, M. J. Gomes, B. Herranz-Blanco, D.
2009, 5, 1575–1581. Liu, J. T. Hirvonen, P. L. Granja, H. A. Santos and B.
86 Y. D. Hu, Q. Wang, J. Y. Wang, J. T. Zhu, H. Wang and Y. J. Sarmento, Nanoscale, 2016, 8, 10706–10713.
Yang, Biomicrofluidics, 2012, 6, 36502. 108 I. M. El-Sherbiny and Y. Abbas, Curr. Pharm. Biotechnol.,
87 M. Windbergs, Y. Zhao, J. Heyman and D. A. Weitz, J. Am. 2016, 17, 673–682.
Chem. Soc., 2013, 135, 7933–7937. 109 S. Lone, S. H. Kim, S. W. Nam, S. Park, J. Joo and I. W.
88 J. Pessi, H. A. Santos, I. Miroshnyk, J. Yliruusi, D. A. Weitz Cheong, Chem. Commun., 2011, 47, 2634–2636.
and S. Mirza, Int. J. Pharm., 2014, 472, 82–87. 110 X. Yu, C. Zhang, S. You, H. Liu, L. Zhang, W. Liu, S.-S. Guo
89 S. Seiffert, J. Thiele, A. R. Abate and D. A. Weitz, J. Am. and X.-Z. Zhao, Appl. Phys. Lett., 2016, 108, 073504.
Chem. Soc., 2010, 132, 6606–6609. 111 Z. H. Nie, W. Li, M. Seo, S. Q. Xu and E. Kumacheva, J. Am.
90 H. Zhang, D. Liu, M. A. Shahbazi, E. M. Mäkilä, B. Herranz- Chem. Soc., 2006, 128, 9408–9412.
Blanco, J. Salonen, J. T. Hirvonen and H. A. Santos, Adv. 112 T. J. Laaksonen, H. M. Laaksonen, J. T. Hirvonen and L.
Mater., 2014, 26, 4497–4503. Murtomaki, Biomaterials, 2009, 30, 1978–1987.
91 W. Li, D. Liu, H. Zhang, A. Correia, E. M. Mäkilä, J. 113 P. M. Valencia, O. C. Farokhzad, R. Karnik and R. Langer,
Salonen, J. T. Hirvonen and H. A. Santos, Acta Biomater., Nat. Nanotechnol., 2012, 7, 623–629.
2017, 48, 238–246. 114 S. Ding, N. Anton, T. F. Vandamme and C. A. Serra, Expert
92 B. Herranz-Blanco, L. R. Arriaga, E. M. Mäkilä, A. Correia, Opin. Drug Delivery, 2016, 13, 1447–1460.
N. Shrestha, S. Mirza, D. A. Weitz, J. Salonen, J. T. Hirvonen 115 I. U. Khan, C. A. Serra, N. Anton and T. F. Vandamme,
and H. A. Santos, Lab Chip, 2014, 14, 1083–1086. Expert Opin. Drug Delivery, 2015, 12, 547–562.
93 F. Kong, X. Zhang, H. B. Zhang, X. M. Qu, D. Chen, M. 116 M. Lu, A. Ozcelik, C. L. Grigsby, Y. Zhao, F. Guo, K. W.
Servos, E. Makila, J. Salonen, H. A. Santos, M. T. Hai and Leong and T. J. Huang, Nano Today, 2016, 11, 778–792.
D. A. Weitz, Adv. Funct. Mater., 2015, 25, 3330–3340. 117 B. K. Johnson and R. K. Prud'homme, Phys. Rev. Lett.,
94 I. U. Khan, C. A. Serra, N. Anton, X. Li, R. Akasov, N. 2003, 91, 118302.
Messaddeq, I. Kraus and T. F. Vandamme, Int. J. Pharm., 118 L. Capretto, D. Carugo, S. Mazzitelli, C. Nastruzzi and X. L.
2014, 473, 239–249. Zhang, Adv. Drug Delivery Rev., 2013, 65, 1496–1532.
95 E. Rondeau and J. J. Cooper-White, Langmuir, 2008, 24, 119 D. Liu, S. Cito, Y. Zhang, C. F. Wang, T. M. Sikanen and
6937–6945. H. A. Santos, Adv. Mater., 2015, 27, 2298–2304.
96 L. H. Hung, S. Y. Teh, J. Jester and A. P. Lee, Lab Chip, 120 D. Liu, H. Zhang, E. M. Mäkilä, J. Fan, B. Herranz-Blanco,
2010, 10, 1820–1825. C. F. Wang, R. Rosa, A. J. Ribeiro, J. Salonen, J. T. Hirvonen
97 E. Amstad, M. Gopinadhan, C. Holtze, C. O. Osuji, M. P. and H. A. Santos, Biomaterials, 2015, 39, 249–259.
Brenner, F. Spaepen and D. A. Weitz, Science, 2015, 349, 121 C. Kunstmann-Olsen, J. D. Hoyland and H. G. Rubahn,
956–960. Microfluid. Nanofluid., 2012, 12, 795–803.
98 O. Gunduz, Z. Ahmad, E. Stride and M. Edirisinghe, Pharm. 122 A. S. Yang, F. C. Chuang, C. K. Chen, M. H. Lee, S. W. Chen,
Res., 2013, 30, 225–237. T. L. Su and Y. C. Yang, Chem. Eng. J., 2015, 263, 444–451.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1881
View Article Online

Tutorial review Lab on a Chip

123 C. C. Hong, J. W. Choi and C. H. Ahn, Lab Chip, 2004, 4, Ferrari, H. R. Allcock, C. M. Sehgal and D. P. Cormode,
109–113. Biomaterials, 2016, 102, 87–97.
124 D. Liu, C. R. Bernuz, J. Fan, W. Li, A. Correia, J. T. Hirvonen 143 M. M. Hasani-Sadrabadi, S. Taranejoo, E.
and H. A. Santos, Adv. Funct. Mater., 2017, 27, 1604508. Dashtimoghadam, G. Bahlakeh, F. S. Majedi, J. J.
125 B. Herranz-Blanco, D. Liu, E. M. Mäkilä, M. A. Shahbazi, E. VanDersarl, M. Janmaleki, F. Sharifi, A. Bertsch, K.
Ginestar, H. Zhang, V. Aseyev, V. Balasubramanian, J. Hourigan, L. Tayebi, P. Renaud and K. I. Jacob, Adv. Mater.,
Salonen, J. T. Hirvonen and H. A. Santos, Adv. Funct. 2016, 28, 4134–4141.
Mater., 2015, 25, 1488–1497. 144 Y. Kim, F. Fay, D. P. Cormode, B. L. Sanchez-Gaytan, J.
126 M. Rhee, P. M. Valencia, M. I. Rodriguez, R. Langer, O. C. Tang, E. J. Hennessy, M. Ma, K. Moore, O. C. Farokhzad,
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

Farokhzad and R. Karnik, Adv. Mater., 2011, 23, H79–H83. E. A. Fisher, W. J. M. Mulder, R. Langer and Z. A. Fayad,
127 P. M. Valencia, P. A. Basto, L. Zhang, M. Rhee, R. Langer, ACS Nano, 2013, 7, 9975–9983.
O. C. Farokhzad and R. Karnik, ACS Nano, 2010, 4, 1671–1679. 145 D. Liu, H. Zhang, S. Cito, J. Fan, E. M. Mäkilä, J. Salonen,
128 N. Kolishetti, S. Dhar, P. M. Valencia, L. Q. Lin, R. Karnik, J. T. Hirvonen, T. M. Sikanen, D. A. Weitz and H. A. Santos,
S. J. Lippard, R. Langer and O. C. Farokhzad, Proc. Natl. Nano Lett., 2017, 17, 606–614.
Acad. Sci. U. S. A., 2010, 107, 17939–17944. 146 Q. Feng, L. Zhang, C. Liu, X. Li, G. Hu, J. Sun and X. Jiang,
129 E. Dashtimoghadam, H. Mirzadeh, F. A. Taromi and B. Biomicrofluidics, 2015, 9, 052604.
Nystrom, Polymer, 2013, 54, 4972–4979. 147 J. Sun, L. Zhang, J. Wang, Q. Feng, D. Liu, Q. Yin, D. Xu, Y.
130 F. S. Majedi, M. M. Hasani-Sadrabadi, J. J. VanDersarl, N. Wei, B. Ding, X. Shi and X. Jiang, Adv. Mater., 2015, 27,
Mokarram, S. Hojjati-Emami, E. Dashtimoghadam, S. 1402–1407.
Bonakdar, M. A. Shokrgozar, A. Bertsch and P. Renaud, 148 I. U. Khan, C. A. Serra, N. Anton, M. Er-Rafik, C. Blanck, M.
Adv. Funct. Mater., 2014, 24, 432–441. Schmutz, I. Kraus, N. Messaddeq, C. Sutter, H. Anton, A. S.
131 F. S. Majedi, M. M. Hasani-Sadrabadi, S. H. Emami, M. A. Klymchenko and T. F. Vandamme, Int. J. Pharm., 2015, 493,
Shokrgozar, J. J. VanDersarl, E. Dashtimoghadam, A. 7–15.
Bertsch and P. Renaud, Lab Chip, 2013, 13, 204–207. 149 J. S. Sun, Y. L. Xianyu, M. M. Li, W. W. Liu, L. Zhang, D. B.
132 M. Russo, P. Bevilacqua, P. A. Netti and E. Torino, Sci. Rep., Liu, C. Liu, G. Q. Hu and X. Y. Jiang, Nanoscale, 2013, 5,
2016, 6, 37906. 5262–5265.
133 L. Capretto, S. Mazzitelli, E. Brognara, I. Lampronti, D. 150 R. Bleul, R. Thiermann, G. U. Marten, M. J. House, T. G. St
Carugo, M. Hill, X. Zhang, R. Gambari and C. Nastruzzi, Pierre, U. O. Hafeli and M. Maskos, Nanoscale, 2013, 5,
Int. J. Nanomed., 2012, 7, 307–324. 11385–11393.
134 M. M. Hasani-Sadrabadi, V. Karimkhani, F. S. Majedi, J. J. 151 R. Thiermann, W. Mueller, A. Montesinos-Castellanos, D.
Van Dersarl, E. Dashtimoghadam, F. Afshar-Taromi, H. Metzke, P. Lob, V. Hessel and M. Maskos, Polymer,
Mirzadeh, A. Bertsch, K. I. Jacob, P. Renaud, F. J. Stadler 2012, 53, 2205–2210.
and I. Kim, Adv. Mater., 2014, 26, 3118–3123. 152 J. M. Lim, N. Bertrand, P. M. Valencia, M. Rhee, R. Langer,
135 R. C. S. Bicudo and M. H. A. Santana, Chem. Eng. Sci., S. Jon, O. C. Farokhzad and R. Karnik, Nanomed.
2012, 84, 134–141. Nanotechnol. Biol. Med., 2014, 10, 401–409.
136 S. Bazban-Shotorbani, E. Dashtimoghadam, A. Karkhaneh, 153 X. J. Kang, C. X. Luo, Q. Wei, C. Y. Xiong, Q. Chen, Y. Chen
M. M. Hasani-Sadrabadi and K. I. Jacob, Langmuir, and Q. Ouyang, Microfluid. Nanofluid., 2013, 15, 337–345.
2016, 32, 4996–5003. 154 J. M. Lim, A. Swami, L. M. Gilson, S. Chopra, S. Choi, J.
137 F. Schutze, B. Stempfle, C. Jungst, D. Woll, A. Zumbusch Wu, R. Langer, R. Karnik and O. C. Farokhzad, ACS Nano,
and S. Mecking, Chem. Commun., 2012, 48, 2104–2106. 2014, 8, 6056–6065.
138 A. J. Mieszawska, Y. Kim, A. Gianella, I. van Rooy, B. Priem, 155 K. I. Min, D. J. Im, H. J. Lee and D. P. Kim, Lab Chip,
M. P. Labarre, C. Ozcan, D. P. Cormode, A. Petrov, R. 2014, 14, 3987–3992.
Langer, O. C. Farokhzad, Z. A. Fayad and W. J. M. Mulder, 156 Y. T. Kim, B. L. Chung, M. M. Ma, W. J. M. Mulder, Z. A.
Bioconjugate Chem., 2013, 24, 1429–1434. Fayad, O. C. Farokhzad and R. Langer, Nano Lett., 2012, 12,
139 M. M. Hasani-Sadrabadi, E. Dashtimoghadam, G. 3587–3591.
Bahlakeh, F. S. Majedi, H. Keshvari, J. J. Van Dersarl, A. 157 P. M. Valencia, E. M. Pridgen, M. Rhee, R. Langer, O. C.
Bertsch, A. Panahifar, P. Renaud, L. Tayebi, M. Mahmoudi Farokhzad and R. Karnik, ACS Nano, 2013, 7,
and K. I. Jacob, Nanomedicine, 2015, 10, 3431–3449. 10671–10680.
140 J. He, Z. J. Wei, L. Wang, Z. Tomova, T. Babu, C. Y. Wang, 158 G. T. Vladisavljevic, N. Khalid, M. A. Neves, T. Kuroiwa, M.
X. J. Han, J. T. Fourkas and Z. H. Nie, Angew. Chem., Int. Nakajima, K. Uemura, S. Ichikawa and I. Kobayashi, Adv.
Ed., 2013, 52, 2463–2468. Drug Delivery Rev., 2013, 65, 1626–1663.
141 L. Wang, Y. J. Liu, J. He, M. J. Hourwitz, Y. L. Yang, J. T. 159 L. Chronopoulou, C. Sparago and C. Palocci, J. Nanopart.
Fourkas, X. J. Han and Z. H. Nie, Small, 2015, 11, Res., 2014, 16, 2703.
3762–3767. 160 F. Fontana, M. A. Shahbazi, D. Liu, H. Zhang, E. M. Mäkilä,
142 R. Cheheltani, R. M. Ezzibdeh, P. Chhour, K. Pulaparthi, J. J. Salonen, J. T. Hirvonen and H. A. Santos, Adv. Mater.,
Kim, M. Jurcova, J. C. Hsu, C. Blundell, H. I. Litt, V. A. 2017, 29, 1603239.

1882 | Lab Chip, 2017, 17, 1856–1883 This journal is © The Royal Society of Chemistry 2017
View Article Online

Lab on a Chip Tutorial review

161 M. B. Romanowsky, A. R. Abate, A. Rotem, C. 164 G. Tishchenko, R. Hilke, W. Albrecht, J. Schauer, K.


Holtze and D. A. Weitz, Lab Chip, 2012, 12, Luetzow, Z. Pientka and M. Bleha, Sep. Purif. Technol.,
802–807. 2003, 30, 57–68.
162 M. L. Eggersdorfer, W. Zheng, S. Nawar, C. Mercandetti, A. 165 I. Limayem, C. Charcosset and H. Fessi, Sep. Purif. Technol.,
Ofner, I. Leibacher, S. Koehler and D. A. Weitz, Lab Chip, 2004, 38, 1–9.
2017, 17, 936–942. 166 D. Quintanar-Guerrero, A. Ganem-Quintanar, E. Allemann,
163 E. Allemann, E. Doelker and R. Gurny, Eur. J. Pharm. H. Fessi and E. Doelker, J. Microencapsulation, 1998, 15,
Biopharm., 1993, 39, 13–18. 107–119.
Published on 28 April 2017. Downloaded by UNIVERSITAT BAYREUTH on 04/07/2017 17:35:53.

This journal is © The Royal Society of Chemistry 2017 Lab Chip, 2017, 17, 1856–1883 | 1883

You might also like