You are on page 1of 31

FEATURE ARTICLE

Drug Delivery www.afm-journal.de

Microfabricated Drug Delivery Devices: Design, Fabrication,


and Applications
Hongbin Zhang,* John K. Jackson, and Mu Chiao*
uniformity of the drug dose. Uncontrolled
Microfabrication technology has enabled the development of novel con- drug release patterns may pose a risk of
trolled-release devices that possess an integration of structural, mechanical, undesirable toxicity and related complica-
and perhaps electronic features, which may address challenges associated tions due to the initial concentration peaks
with conventional delivery systems. In this feature article, microfabricated arising from burst release and low efficacy
because of subsequent rapid drug con-
devices are described in terms of materials, mechanical design, working prin-
centration decline below the therapeutic
ciples, and fabrication methods, all of which are key features for production range.[2]
of multifunctional, highly effective drug delivery systems. In addition, the cur- In the 1960s, the concept of sustained
rent status and future prospects of different types of microfabricated devices release came up for achieving zero-
for controlled drug delivery are summarized and analyzed with an emphasis order release kinetics.[3] Thereafter, some
on various routes of administration including ocular, oral, transdermal, and polymer-based matrices and membrane-
controlled vehicles have been developed
implantable systems. It is likely that microfabrication technology will continue
to realize a constant release rate. Although
to offer new, alternative solutions to design advanced and sophisticated drug sustained release systems, to some extent,
delivery devices that promise to significantly improve medical care. reduce the risk of toxic effects by attenu-
ating the peaks and valleys of plasma drug
concentration, most of them release drugs
1. Introduction over a limited time period (less than a day) and their release
rates may be influenced by environmental conditions, such
Drug delivery, the method by which therapeutic agents are intro- as pH, temperature, and ionic strength, which lead to unde-
duced into the body, plays a crucial role in achieving optimal sired variation in efficacy.[4–6] On the other hand, many clinical
treatment efficacy of medications and provides improvements situations actually need more than a zero-order drug release,
for drug safety, convenience, and patient compliance. With an requiring an on-demand control of drugs which may vary with
increasing awareness of the significance of pharmacokinetics time or patients’ conditions. Moreover, the emergence of biop-
and biodistribution for the successful therapeutic effect of a harmaceuticals including proteins, peptides, vaccines, etc., also
drug, the optimal application of drug delivery technologies has calls for new delivery strategies due to their low bioavailability
evolved substantially over time.[1] and poor stability in the gastrointestinal tract, making them not
Primitive methods of delivering drugs were mainly through suitable to be delivered by oral methods.[4,7]
physical digestion of chewed medical plants, soot inhala- Therefore, new advanced delivery systems capable of targeted
tion from the burning of medical substances, or application dosing with an adjustable rate for a definite time period are
of mashed herbs on skin. When the effective ingredients of needed. To date, with advances in materials science, chemi­cal
such materials could be synthesized or extracted from plants engineering and manufacturing technology, numerous novel
sources, a variety of pharmaceutical formulations were pro- controlled release systems such as nano-/microcarriers,
duced using different excipients which include tablets, cap- poly­mer/hydrogel matrices, and microfabricated devices/
sules, syrups, emulsions, ointments, solutions, and so on.[2] implants, have sprung up.[1,8,9] The existing drug delivery sys-
Drug delivery was extended to include intravenous and intra- tems can roughly be categorized into two types: nonstimulated
muscular injections, eye drops, implants, suppositories, etc. release such as diffusion from polymers or stimulated release
However, these traditional delivery systems are simple and usu- such as from mechanical devices. The nonstimulated group is
ally follow first-order release kinetics, lacking consistency and mainly based on the development of new materials by chemical
synthesis and biological techniques. In these systems, materials
act as drug depots from which the drug release depends on the
Dr. H. Zhang, Prof. M. Chiao
Department of Mechanical Engineering
material’s chemical/physical properties and structures.[10–12]
The University of British Columbia For example, drug release from polymeric materials generally
Vancouver, BC V6T 1Z4, Canada is controlled by mechanisms of diffusion, chemical reaction-
E-mail: izhanghongbin@gmail.com; muchiao@mech.ubc.ca related polymer degradation or chemical bond cleavage of
J. K. Jackson the drug from polymer chains, and solvent activation such as
Department of Pharmaceutical Sciences swelling or osmotic effects.[13,14] Nanomaterials usually control
The University of British Columbia
Vancouver, BC V6T 1Z3, Canada release drugs by physically or chemically responsive disintegra-
tion, and targeting delivery via the incorporation of biological
DOI: 10.1002/adfm.201703606 agents.[15] The stimulated group of delivery systems includes

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (1 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

controlled release devices that involve materials, mechanical


design, and fabrication techniques, such as micro-electrome- Hongbin Zhang is currently
chanical systems (MEMS) and nano-electromechanical systems a postdoctoral research
(NEMS)-based devices.[16–19] The devices consist of different fellow in the Department
functional components such as micropumps, microvalves, of Mechanical Engineering,
reservoirs, channels, etc., forming an integrated system for University of British Columbia
achieving more sophisticated and precise drug delivery.[17,20] (Canada). He received his
Although many types of devices are still in different stages of B.S. degree from the Sichuan
production and clinical evaluation, the development of the con- University (China) and Ph.D.
trolled release devices has shown great potential to offer new from the University of Science
ways of drug administration with effective therapeutic regi- and Technology Beijing
mens.[18] The fast evolution of the microfabricated drug delivery (China). His current research
devices is reflected in Figure 1A, where the number of relevant interests focus on soft mate-
publications per year from 2000 to 2016 is presented and the rials, controlled drug delivery systems, surface modifica-
proportion of publications has reached around one third in the tion and antifouling, and tissue engineering.
annual publications on drug delivery systems.
The purpose of this feature article is to provide a comprehen- John K. Jackson is a research
sive overview of the emerging controlled drug release microfab- scientist in the Faculty of
ricated devices. The devices are first discussed from the aspects Pharmaceutical Sciences
of materials, design principles, and manufacturing technolo- at the University of British
gies, followed by their respective reviews on the applications Columbia. He has worked on
according to their different features and various administration polymeric controlled release
routes for drug delivery. drug delivery systems for
over 25 years. His interests
include drug coating of
medical devices and he was
2. Microfabricated Drug Delivery Devices the first to coat implantable
Many types of devices have been designed and fabricated for stents with drugs such as
controlled drug delivery over the years. Unlike traditional drug paclitaxel.
delivery systems which mostly are composed of drugs together
Mu Chiao is a professor
with excipients or carrier materials, drug delivery devices are
in the Department of
fabricated through a combination way with different materials,
Mechanical Engineering at the
mechanical design, and specific manufacture technologies
University of British Columbia
(Figure 1B).[21] This provides variety in release mechanisms that
(Canada). He obtained his
are independent of material properties to achieve more accu-
B.S. degree from the National
rate and active control over drug delivery.
Taiwan University (Taiwan)
and Ph.D. from the University
of California–Berkeley (USA).
2.1. Materials
His research interests include
microelectromechanical
Material selection is the process of seeking the material with
systems (MEMS), BioMEMS/
appropriate properties to meet design requirements, serve
drug delivery, MEMS/protein interaction, and micro-optical
device performance goals, and minimize cost. Some aspects
scanner for biological applications.
to be considered include mechanical properties for processing
and use, stability when loaded with drugs both in storage and
in physiological environments, biosafety to avoid the genera- immersion in a chemical solution (wet etching) or by exposure
tion of toxic substances especially for implantable devices, etc. to chemically reactive plasma (dry etching). For example, fast
anisotropic etching of single-crystal silicon can be achieved
in strong base solutions, which allows fabrication of a large
2.1.1. Rigid Materials variety of silicon structures in a controllable and reproducible
manner. In addition, photolithography and chemical/physical
Silicon is one of the initially used materials in microfabricated vapor deposition can also be used for the silicon material in
devices because of its favorable electrical and mechanical prop- microfabrication.
erties and easy integration with conventional semiconductor Silicon is chemically and biologically stable, which is suit-
processing techniques.[22] This material could be a variety of able for drug loading and delivery. And importantly, silicon
thin single-crystal wafers (0.25–10 mm) with different crys- has shown to have biocompatibility and nonimmunogenicity
talline orientations. Various microstructure or micropatterns approximately equivalent to tissue culture polystyrene on
such as microchannels and micropores are able to be cre- either its flat or micro-/nanostructured surface.[22–26] These
ated by etching directly into the silicon substrate either by features make silicon a good material for making components

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (2 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

of drug delivery. It can be used alone or be bonded to silicon


using anodic bonding or silicone rubbers to form complex sys-
tems with different microstructures (e.g., microfluidic chips)
for controlled drug delivery.[42–44] Porous inorganic materials
such as mesoporous silica[45,46] and zeolite[47] in different
shapes have attracted special attention in drug delivery micro-
devices due to the large surface area and pore volume.[48,49] In
addition, functionalization of the porous materials with mole­
cular, supramolecular, or polymer moieties enable fabrication
of new devices with great versatility in controlled drug delivery
utilizations including sustained release, targeted release, and
stimuli-responsive release.[50,51] Other rigid materials such as
metals,[52–54] ceramics,[55,56] silicon nitride,[57,58] and polycrystal-
line silicon[59,60] have also been used in developing various drug
delivery devices.

2.1.2. Polymers

Polymers have been used extensively in biomedical applications


including controlled drug delivery systems. Much of the con-
trolled-release technology relies on polymer properties such as
stimuli-responsive behavior, degradability, tissue-like mechan-
ical strength, controllable hydrophilicity and hydrophobicity,
and tunable structures. However, the current methodology
used for polymers sometimes is not efficient enough to provide
the high-precision control over the release timing and rate that
is often required for controlled drug delivery. With the develop-
ment of microfabrication technologies, polymers show superior
properties to silicon for bioMEMS applications with regard to
the low cost and controllability over various physicochemical
properties.
Poly(dimethylsiloxane) (PDMS) is a commercially avail-
Figure 1.  A) Microfabricated drug delivery device documents published
per year from 2000 to 2016 and its proportion in the annual publications able polymer for the production of biomedical devices such
on drug delivery systems. The presented data collected from the Web of as microfluidic chips, drug delivery devices, and surgical
Knowledge by using the key search keywords of “drug delivery,” “device,” implants.[61–63] Liquid PDMS prepolymers can be cured by
and “micro-,” or “drug delivery system.” B) Influencing factors of a micro- thermal crosslinking with crosslinkers at temperatures less
fabricated device for controlled drug delivery. than 100 °C or even at room temperature. The exposure to high
energy irradiation or the use of platinum as catalyst can also
of drug delivery systems. So far, silicon has been processed be the choice of curing methods for PDMS, making the mate-
into platforms with different microstructures in drug delivery rials easy to handle. Additionally, PDMS is chemically inert, gas
devices such as microneedles (MNs),[27,28] microreservoir- permeable, isotropic, and optically transparent down to about
based devices,[29,30] and nanoporous and nanochannel micro- 300 nm, allowing UV crosslinking of moldable prepolymers in
chips.[31,32] Microstructured silicon can be used as a template as photolithographic patterning. The inherent mechanical flexi-
well for the formation of microneedles or microchannels from bility of PDMS can be beneficial to construction of micropumps
other materials, with the patterns being transferred to the end and microvalves that operate by inducing deformations in the
state material through methods such as micromolding.[33,34] substrate itself. Nevertheless, PDMS-based devices may have
Great potential has been shown for silicon-based devices as biofouling problems from nonspecific protein/hydrophobic
an essential platform for future diagnostics/therapeutics due analyte adsorption in the physiological environment owing to
to the ability to integrate electronic components directly within the intrinsic hydrophobicity of PDMS.[64] Thus, surface modifi-
microfluidic and drug delivery systems.[35,36] Although expen- cation and treatment may be needed to ensure the performance
sive microfabrication techniques and clean rooms are required of implantable PDMS devices.
in the fabrication of silicon-based microsystems, much of the Other polymers such as polymethylmethacrylate (PMMA),
added cost and complexity associated with silicon processing polyethyleneglycol, poly(lactic acid) (PLA), polyglycolic acid,
can be compensated by the economics of scale associated with poly(d,l-lactide-co-glycolide) (PLGA), poly(caprolactone) (PCL),
miniaturization. poly(glycerol-sebacate), polyurethanes, and SU-8 are being
Glass is another common material which can be microma- investigated as alternative materials in the fabrication of drug
chined by lithography and etching.[22] Glass-based micronee- delivery devices.[22,65,66] For example, micro-mucoadhesive
dles,[37] fibers,[38–40] and tubes[41] are examples in the application devices (≈100 µm) have been micromachined from PMMA

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (3 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

to deliver drug to the wall of the intestine through the oral temperature, biological molecules, light, electromagnetic fields.
route.[67] In addition, multipulse drug delivery from a resorb- When choosing responsive materials to fabricate drug delivery
able polymeric microchip device made of PLA reservoirs and devices, the sensitivity to stimuli, response speed, extent, and
PLGA membranes was demonstrated.[68] stability of the material are key factors determining the perfor-
For polymers, replication techniques such as soft lithography mance of products. Integration of stimuli-responsive materials
and direct plastic micromachining are applicable for microfab- in the devices with careful and creative design could be a very
rication, allowing repeated production of many devices with promising approach toward smart on-demand drug delivery
a single master to decrease cost. Many traditional manufac- devices. More materials and responsive mechanisms are yet to
turing techniques for polymers, such as injection molding and be discovered to meet the emerging needs in the field of device
embossing, can also be applied in the microfabrication field at development.
a much smaller scale, which provide more design flexibility.
In regard to the immunogenicity-related safety issue, many of
the polymer surfaces, including those that are microstructured, 2.2. Design Considerations/Principles
do not elicit a significant immune response. All of these make
polymers suitable for microfabricated drug delivery devices. Numerous drug delivery devices with different shapes, sizes,
functions, and drug release control mechanisms have been
developed to serve different routes of drug administration and
2.1.3. Relationship between Material Properties/Functionality enhance the control over drug delivery behaviors. To achieve
and Device Design these objectives, a rational design for the device is a crucial step,
where a number of factors including the way of drug loading/
In the design and fabrication of drug delivery devices, the release and the specific area where the drug will be released
matched structures and properties/functionality of device should be considered.
materials with the carried drugs, play a key role in optimizing
performance for each specific application besides basic consid-
erations of safety, availability, and cost. For example, good drug– 2.2.1. Reservoir Design
drug carrier compatibility may increase the stability of drugs
in devices and prolong the storage period. Proper mechanical Matrix type delivery systems where drugs are dissolved, dis-
strength of device helps to maintain the structural integrity of persed, or chemically bonded, form a resistant support to disin-
devices for robust controlled drug delivery. The integrability tegration for delay and control of the drug release. However, the
and potential interactions of different components for a device matrix systems may face limitations such as insufficient drug
should be also taken into consideration in the device design loading capabilities, burst release, and problems associated
step. Among the various properties of materials, degradability with the biodegradability and metabolism of the matrix after
and stimuli-responsive aspects are important in the fabrication degradation. In contrast, reservoir-based delivery devices which
of drug delivery devices. contain cavities for loading drug, can offer an isolated space of
Degradable materials including metallic materials such as a large volume to protect drugs from the outside environment
magnesium alloys[69–71] and polymers[72–74] could be used as and allow drugs to be loaded in solid, liquid, or gel state.[89] By
the matrix, coatings, or functional components of a device to utilizing microtechnologies, these devices are able to be fabri-
control drug release.[75–77] Preferably, to be the matrix material cated in a smaller overall size or profile, which can incorporate
(mostly for implantable devices), the material should maintain features or mechanisms that enable more precise adjustment
optimal mechanical properties until the loaded drug is released of drug delivery rate and provide ways to reach difficult-to-treat
and then degrade, being innocuously absorbed and excreted locations.[89]
by the body. As coatings, the degradation time of materials Through proper designs of the reservoirs, some other
should match the needs of specific applications, such as to pre- functions can be imparted to the device. For example, highly
vent devices from damage or premature drug release before localized and unidirectional release of drugs was achieved with
reaching the target area. To perform well as functional compo- a reservoir-containing microdevice.[90] The devices consisted
nents for drug release, the materials should have tunable and/ of a planar semiclosed reservoir made of drug-impermeable
or controllable degradation speeds. For example, as a sealing materials. The top face (the reservoir opening) of the device was
membrane for drug release outlets, degradation of the mem- chemically modified to graft protein or other types of ligands
brane might be triggered by specific chemical or biological trig- for the bioadhesive purpose. Because of the asymmetric struc-
gers such as ions, pH, enzymes, etc., or controlled through tai- ture and surface modification, the device could attach and
loring the material structures. asymmetrically deliver drugs (especially peptides and proteins)
Stimuli-responsive materials capable of regulating transport/ to the intestinal mucosa in the gastrointestinal (GI) system
discharge of drug molecules by shape deformation, movement, after oral administration. The flat reservoir design not only
or assembling/disassembling on receiving external signals are increased contact area of the device with the intestinal wall to
attractive for controlled drug delivery devices.[78–81] They could enhance the adhesion ability, but also minimized shear distur-
be fabricated into different functional components for a device bances from the flow of liquids through the intestine. The drug
such as valves,[82–84] gating membranes,[51,85] channels, and impermeable walls and base of the device effectively limited the
hinges[86,87] or serve as drug depots,[79,88] which can trigger and leakage and exposure of bioactive drugs to enzymatic degrada-
stop drug flux in response to environmental stimuli like pH, tion in the intestinal lumen.[90]

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (4 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 2.  Examples of single-reservoir and multireservoir devices for drug delivery. A) An electrochemically driven microfluidic drug delivery device.[29]
(a) The electric potential is applied between top (gold membrane) and bottom electrodes; (b) Two main electrochemical reactions occur: dissolution
of the gold membrane and electrolysis of water resulting in gas release; (c) The generated microbubbles propel drug solution out; (d) The reaction
continues until fluid transport stops. Reproduced with permission.[29] Copyright 2009, Springer. B) A biodegradable multireservoir device.[68] (a) Dia-
gram of the polymeric device; (b) Pulsatile release of drugs from polymeric device in vitro. Reproduced with permission.[68] Copyright 2003, Nature
Publishing Group.

The drug-containing reservoir can also be totally closed until 2.2.2. Release Mechanism Selection
signals are given to remove the sealing membranes. Such design
reduces the dependence on the external physiological conditions The core of a drug delivery device is the method for controlling
and provides a pulsatile fashion of drug delivery. For example, in drug release behaviors to meet different requirements in real
an electrochemically driven device, the reservoir is sealed with applications. There exist different control mechanisms for drug
a gold membrane (Figure 2A).[29] When an electrical potential release, which have been proposed and undergoing investiga-
is applied between upper (the gold membrane) and lower elec- tion for drug delivery devices. Here, three main categories of
trodes, the capping membrane dissolves and releases drug. The controlled release mechanism are discussed.
drug release is able to be accelerated by propelling of microbub- Diffusion Controlled Release: Diffusion controlled systems
bles generated from water electrolysis inside the device. rely on diffusion of drug out of or through a membrane or
A device can be designed to have a single reservoir or mul- matrix that may be nonporous or microporous.[92] The rate
tiple reservoirs. Single-reservoir devices provide more room for determining step is the transport rate of drugs through the
drug loading which is beneficial to the long-term use, while microstructured media which work as barriers to prevent fast
multireservoir devices can store drugs separately, allowing for diffusion of solutes from a region of high concentration (reser-
incorporation of different drugs in one single device for syner- voir) to a region of low concentration (environment). By using
gistic treatment of diseases such as cancer. Individual control polymeric semipermeable membranes or crosslinked gels with
over drug release from each separate reservoir of the multires- differential pores, density, and erosion rates, sustained drug
ervoir device may be achieved by properly choosing the release diffusion was demonstrated in some devices such as ocular
mechanism, creating the possibility of achieving many com- implants for intravitreous drug delivery and drug-eluting stents
plex release patterns. Accordingly, electrochemically controlled for intravascular therapies.[93]
release was demonstrated successfully in multireservoir devices Silicon microchannels with different geometries have been
for multidose drug delivery.[91] Another example is based on designed to regulate the passive diffusion of molecules. As
polymer-degradation controlled mechanism, where each res- shown in Figure 3A, the simulation of thermal diffusivity in
ervoir is sealed by degradable membranes made of PLGA of six different microchannel configurations shows that drug dif-
different molecular weights (Figure 2B).[68] These membranes fusion in each microchannel exhibits different characteristics,
exhibit different degradation speeds, resulting in pulsatile drug giving rise to different diffusion rates.[94] As the channel width
release for different drugs over a period of months. In addition, decreases to approach the hydrodynamic diameter of the solute,
the release characteristics are possible to be tailored for specific linear non-Fickian behavior of solutes could be achieved. This
applications by simply varying the parameters of the device res- could be observed in other microporous media such as zeolites,
ervoirs (e.g., numbers, volumes) and membranes (e.g., thick- which is considered to be a result of traffic control phenomena,
ness, molecular mass, materials). called single-file diffusion and/or pore wall drag effects via

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (5 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 3.  Mechanisms of controlled release. A) Simulations of drug diffusion through different microchannel configurations (the pseudocolor images
indicate the local concentration of the drugs). Reproduced with permission.[94] Copyright 2012, Hindawi Publishing Corporation. B) Reciprocating
micropump operation. (a) Schematic configuration of a typical reciprocating pump; (b) Drug release during the pumping mode; (c) Solution refilling
during the supply mode. C) Schematic description of an osmotic drug delivery device. D) Schematic illustration of a magnetic micropump and its
operation.[104,105] Reproduced with permission.[105] Copyright 2011, Royal Society of Chemistry.

molecular interaction. By precisely tailoring the size, length, be controlled externally by patients or operators would be more
density, and surface composition of nanochannels according to favorable.[92] The direct method for pulsatile release is to open
the physicochemical characteristics of the biological molecule and/or close the drug release outlet of the device actively. As
of interest, zero-order long-term drug delivery is achievable for mentioned above, the dissolution of sealing membranes by elec-
a wide variety of drug classes. trochemical/electrothermal reactions allows for the triggered
Outlet Valve Controlled Release: In addition to sustained release of enclosed drugs from each reservoir (Figure 2A).[29,95]
release systems, efforts to improve control over the rate and Although the drug release following removal of the membrane
extent of drug delivery currently center around the design of is through diffusion, the rate determining step depends on the
triggered-release systems by external signals. In some situ- opening of the drug reservoir. This capability of controlling the
ations such as insulin delivery for diabetes, sustained release outlet opening and/or closing of a device, which named outlet
may not be the optimal fashion, while pulsatile release that can valve control mechanism, plays a pivotal role in accomplishing

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (6 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

the triggered release. Moreover, by blocking and unblocking the have simple structures, their actuation mechanisms are rarely
micro-orifices of reservoirs with magnetic particles or stimuli- used in drug delivery devices because of their relatively lower
responsive microgels, reversible control of the outlet valve (on/ driving effect and performance (e.g., flow rate, response
off) can be obtained to repeatedly trigger drug release from a time, and pressure generation) compared to the mechanical
single-reservoir device until the drug is exhausted.[96,97] actuation.
Micropump Driven Release: Micropumps are an essential Micropumps provide the potential to achieve complex drug
component in fluid transport systems to precisely meter and release patterns, but the limitation may be that they are com-
control the motion of liquids.[98] Integration of a micropump monly only suitable to administer drug solutions or suspen-
in a delivery device offers a number of options to drive drug sions, which should be considered when designing a drug
release in a controlled manner and achieve more complex delivery device. Some other basic requirements must be
release patterns.[65,99] Micropumps can basically be categorized thought about when designing and choosing micropumps for
into two types: mechanical and nonmechanical, depending on drug delivery applications, which include drug biocompatibility,
the presence of moving parts.[100] The movement mechanisms actuation safety, desired and controllable flow rate, small size,
for mechanical pumps usually include reciprocating, rotatory, and low power consumption.[107]
or peristaltic, in which the reciprocating motion is the most
commonly used in drug delivery systems.[101] The recipro-
cating micropump normally is comprised of a pump chamber 2.2.3. Functional Structure and Appearance
equipped with an actuator and a flexible diaphragm membrane,
and microchannels and microvalves to control fluid flow.[99] The For versatility, acceptability, and achievement of functions,
actuator converts energy into the movement or deformation in some other factors are involved in designing the device.
a to-and-fro motion of the membrane to provide force to fluid The shape and overall device size are major considerations.
flow out and into micropumps. The movement of the mem- The implantable device should be small enough to allow for
brane generates a positive pressure within the pump chamber, intraocular or subcutaneous implantation in a doctor’s office
resulting in opening of the outlet valve for delivery of the drug with a profile that is not obtrusive or apparent. For example,
to the external environment from the pump chamber. Subse- an intravitreal insert marked as Iluvien is designed as a cylin-
quently, the membrane moves back to the original position drical shape (Figure 4A), and miniaturized to allow for injection
creating a negative pressure to close the outlet valve but force using a proprietary 25-gauge injector system with a minutes-
the inlet valve to open to allow the drug supplement by flowing long in-office procedure.[108] Another cylindrical magneti-
into the chamber. Figure 3B shows the schematic diagram cally actuated device was also reported for minimally invasive
of the mechanical pump and its functioning.[99,101] Various implantation by a 12-gauge needle for prostate cancer treat-
mechanisms have been used to develop different actuators for ment (Figure 4B).[109] The appearance of the device should
mechanical micropumps, including piezoelectric, electrostatic, be designed and fabricated according to the characteristics of
thermopneumatic, bimetallic, shape memory alloy, ionic con- specific target organs such as devices for installing in or on
ducting polymer films, and so on.[100] These actuators take eye balls or in internal acoustic meatus. For some devices, the
energy from electricity, heat, liquid pressure, or air pressure, appearance itself serves as a functional structure for achieving
and convert it into some kind of mechanical motion. Thus, they better performance. A typical example is microneedles, the
normally function with the aid of a power supply such as bat- appearance of which describes functions. The needle has to be
teries that needs to be integrated with the micropump. sharp to pierce through the stratum corneum of the skin and its
There are mechanical micropumps with no need of an on- length has to be short enough to avoid stimulating the nerves
board power supply, such as osmotic pumps and magnetic in the dermis.[110] Moreover, the main part of the microneedle
pumps. Osmotic-type micropumps usually have two chambers can also be used as a drug reservoir.[20] Another example is
(Figure 3C).[100,102] One chamber serves as a drug reservoir con- punctal plugs for drug delivery to the eye by inserting into the
taining drug solutions and the other chamber which is made lid punctal (Figure 4C).[111] Various punctal plug designs with
up of a semipermeable membrane contains osmotic driving different appearances were proposed by Eagle Vision, as shown
agents. These chambers are separated from each other by a in Figure 4C(c).[111] They have been claimed to provide unique
flexible membrane or a piston. In operation, the water mole- advantages for applications, including improved patient com-
cules diffuse through the semipermeable membrane, resulting fort, prolonged retention and easy insertion operation, etc.
in the volume expansion of the osmotic engine chamber. The For MEMS-based drug delivery devices, several subsystems are
expansion drives a piston forward and expels the drug solu- usually required to provide all necessary functions, which include
tion out through an orifice. Magnetic pumps generate mechan- a power source, wireless communication, and control circuitry
ical movement of a piston[103] or deformation of a membrane that programs the rate and amount of each dose released from
(Figure 3D)[104,105] using external magnetic fields which allow reservoirs in the device. These subsystems may create a design
for independent control over the actuation process, and hence hurdle for incorporating a sufficient quantity of drug in the device.
the drug delivery rate and timing. Therefore, an optimal design that can separate different working
Unlike mechanical micropumps, nonmechanical ones do components of a device and distribute the components in various
not require moving parts, instead they generate flow through a places may be desired and suitable candidate drugs might be
number of nonmechanical energies including self-diffusiopho- potent and can be formulated in high-concentration forms. Oth-
resis, electrophoresis, bubble propulsion, and the generation of erwise, a refillable design and structure should be considered to
density gradients.[106] Although nonmechanical micropumps prolong the use of device for long-term dosing regimens.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (7 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 4.  A) Schematic representation of intravitreal insertion of Iluvien via an injector. B) A minimally invasive drug delivery device: (a) Schematic and
(b) image of the actual device. Reproduced with permission.[109] Copyright 2015, Royal Society of Chemistry. C) (a) Schematic illustration of a punctal
plug delivery device; (b) Schematic illustration of punctum location (inset) and placement of punctual plugs in the punctum of the eye; (c) Schematic
representation of assorted designs of silicone-based punctual plugs and each design has been claimed to provide a unique advantage to the dry eye
syndrome (DES) patients. Reproduced with permission.[111] Copyright 2015, Elsevier.

2.3. Fabrication Technologies tronics industry. These techniques may involve a series of
processing steps: thin-film deposition, lithography, etching,
Advances in new micro- and nanotechnologies have been and substrate bonding, which allow for transferring and con-
changing the way of drug delivery and enabled fabrication of structing microscale features on the surface of a desired sub-
novel devices to deliver drug efficiently and precisely to target strate or into a bulk material. In fabrication of microdrug
regions. The use of microfabrication techniques allows for the delivery devices, the most commonly applied technique is
accurate control over devices’ surface microarchitecture, topog- photo­lithography.[114] It can transfer a computer-generated
raphy, and feature size, which play an important role in the pattern from a mask to a substrate (silicon, glass, etc.) via the
device fabrication and function accomplishment.[112,113] localization of light, as shown in Figure 5A(a).[112,115] The sub-
strate is coated with a photosensitive resist material (positive in
the figure), followed by exposure to UV light through a pho-
2.3.1. MEMS/NEMS Fabrication Technology tomask with the pattern to be transferred. After that, the sub-
strate is immersed in a developer solution, which selectively
Basic microfabrication techniques for the manufacture of removes UV-exposed regions, resulting in a high-fidelity rep-
MEMS- or NEMS-based devices are developed from the elec- lica of the mask image on the substrate surface. Subsequent

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (8 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 5.  A) Illustration of various micromachining processes. (a) Photolithography-based microfabrication process; (b) “Soft lithography” process
for polymer-based devices. B) Summary of the microfabrication techniques. C) Graphical examples of 3D printed tablets with various designs and
functionality.[118] Reproduced with permission.[118] Copyright 2016, Springer.

micromachining processes could be applied to either selectively ymer–silicon/glass) is one of the most important fabrication
remove or deposit material in the patterned regions. Microma- techniques in microsystem technology, which may involve
chining techniques can be categorized into surface microma- fusion bonding and electrostatic bonding. It can be used for
chining and bulk micromachining.[115] Surface micromachining packaging and encapsulation, or to construct complex 3D struc-
is an additive fabrication method by chemical or physical tures as a functional unit in the final microsystem.
deposition. Bulk micromachining, in contrast, is a subtractive There are other techniques developed for microfabrication,
method by wet or dry etching. Since the photolithography is some of which are summarized in Figure 5B.[113,117]
limited by the wavelength of the light used for exposure, high
energy lithographic techniques including X-ray LIGA (lithog-
raphy, electroforming, and molding), e-beam lithography, and 2.3.2. 3D Printing Technology
ion-beam lithography have been developed to fabricate finer
structures in the nanometer scale. In recent years, 3D printing, a rapid additive manufacturing
A second pattern transfer approach called soft lithography refers method has started to demonstrate fine enough resolutions
to a group of techniques using elastomeric stamps and molds.[114–116] in microfabrication via direct printing of structures. This
Typically, an elastomer (such as PDMS) is patterned by curing approach includes a wide variety of manufacturing techniques
on a conventional micromachined rigid mold (Figure 5A(b)),[112] such as stereolithographic, powder-based, selective laser sin-
which enables rapid prototyping of similar structures. In addi- tering, fused deposition modeling, and semisolid extrusion 3D
tion, the mold can be repeatedly used, which reduces the cost printing, which are all based on computer-controlled depositing
considerably. Substrate bonding of different materials (silicon– of materials (layer-by-layer) to create freeform and complex
silicon, silicon–glass, glass–glass, polymer–polymer, and pol- structures.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (9 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

3D printing may enable direct fabrication of personalized and elevation of intraocular pressure, result in poor patient
drug delivery devices with tailored functionality, due to its compliance.[130] Because of the limitations associated with
precise spatial control over the device geometry including size, the current methods of ocular drug delivery, attention has
shape, and internal structure (Figure 5C).[118] The printed device turned to developing novel therapeutic strategies and new sys-
may enhance the stability of multiple drugs with a tablet, ensure tems such as microfabricated devices for high drug delivery
precise dosing for each drug, and regulate release kinetics of efficiency.[129,133–136]
each component by incorporating well designed structures that
can modulate dissolution and diffusion profiles.[119] 3D printing
technology brings us a step closer to customized medicine and 3.1.1. Contact Lenses
makes the automated and high-speed manufacture of various
complex structures possible for microfabricated drug delivery Contact lenses usually composed of poly(2-hydroxyethyl meth-
systems in future.[120,121] acrylate) (pHEMA)[137,138] or silicone hydrogels[139] are com-
monly used today for vision correction and present an attractive
platform for ocular drug delivery because they are noninva-
3. Microfabricated Devices for Different sive, replaceable, and well accepted by patients. Although the
drug-containing contact lenses are still in preclinical stages,
Administration Routes and Disease Models
in vitro testing has shown better drug bioavailability than the
Biomedical researchers have attempted to explore different topical eye drops due to reduced tear mixing between the lens
devices capable of drug administration through essentially and the cornea, which prolongs the retention time of drugs.
every portion of the body in order to achieve the convenient The challenges for the contact lens-based drug delivery devices
but efficient treatment. According to specific physiological are to increase drug loading amounts and to achieve sustained
structures, features, and functions of different tissues, organs, drug delivery over a long time period. Approaches including
and systems in body, various devices have been engineered and adjusting the component ratios of lens and incorporating drug
fabricated to adapt to and meet requirements of different drug encapsulated nanoparticles or liposomes in the lens matrix have
administration routes in applications.[122–124] The representative shown varying degrees of promise in lengthening sustained
samples in this section are summarized in Table 1. release of drug (days to weeks). A recent study reveals that by
loading vitamin E or vitamin A could remarkably increase the
loading of hydrophilic glaucoma drugs (timolol and brimoni-
3.1. Ocular Delivery dine), but their release duration time was not significantly
altered.[137] In another example, sustained zero-order release of
Intraocular diseases such as glaucoma, retinitis pigmentosa, drugs was achieved by modifying contact lens designs.[140,141] In
age-related macular degeneration (AMD), and diabetic retin- this special design, a drug-containing annular film made with
opathy, are the leading causes of serious vision loss around the degradable poly(lactic-co-glycolic) acid is created as a drug depot.
world.[125–128] However, effective drug delivery to ocular tissues Then, the film depot is coated in the nondegradable pHEMA
remains a major challenge to pharmacologists and scientists hydrogel to form the lens. These lenses showed sustained drug
due to the unique anatomy and physiology of the eye. Struc- delivery with zero-order kinetics for up to four weeks and the
tural layers of ocular tissue (cornea, sclera, conjunctiva, etc.), drug release rate can be tuned by changing the proportion of
tears and blood–ocular barriers (blood aqueous and blood–ret- drugs in the PLGA film.[140,141] Limitations of the usage of con-
inal barriers) represent the protective barriers restricting the tact lenses as drug delivery devices include the necessary long
entry of drugs to the target sites such as the ciliary body, retina, wear time that may raise safety concerns (e.g., corneal abrasion
and choroid.[129–131] and infection), and possible drug leaching out of the lens due
Conventional ocular drug delivery therapies including topical to the hydrated storing environment for lenses.
eye drops, oral medications, and intraocular injections, are lim-
ited in efficacy. For example, eye drops that account for 90%
of currently available ophthalmic formulations suffer very low 3.1.2. Iontophoretic Devices
intraocular bioavailability of around 5% owing to rapid dilution
by tears and require frequent administration.[132] Ointments, Iontophoresis is another noninvasive technique for ocular
gels, and liposomes have been used in eye drops to prolong drug delivery, in which a mild direct electric current is applied
drug contact time with the cornea and enhance drug delivery, to enhance penetration of charged drugs across the sclera/
but only limited increases in intraocular bioavailability (about cornea and into the choroid, retina, and vitreous.[142,143] The
10%) can be reached, since most of the drug is cleared by the iontophoretic device generally is composed of a direct cur-
local systemic absorption. Drug delivery via oral routes needs rent power source and two electrodes. To deliver the drug, an
large systemic doses to ensure adequate intraocular thera- electrode carrying the substances with the same charge as the
peutic levels after passing through the blood–retinal barrier. electrode is applied at the target area, and the ground elec-
This is often associated with significant side effects. Intravit- trode is placed elsewhere on the body to complete the circuit.
real injections provide a direct approach to delivering drugs to The drugs then are driven into the tissue by electrorepul-
ocular tissues, especially those in the posterior segment. How- sion at either the anode (for positive drug) or the cathode
ever, repeated injections and potential complications including (for negatively charged drug).[144] The majority of the current
cataract formation, vitreous hemorrhage, endophthalmitis, ocular iontophoretic devices use a relatively large eye cup or

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (10 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Table 1.  A summary of different drug administration routes described in the text.

Administration Device categories Characteristics Ref.


routes
Ocular delivery Contact lenses pHEMA-hydrogel contact lenses with increased load of hydrophilic glaucoma drug [137]
Sustained zero-order release with a degradable drug-loaded annular film [140,141]
Iontophoretic devices Delivering charged drugs into the vitreous cavity through the sclera without tissue damage [145]
Punctal plugs Tiny biocompatible lacrimal plugs with different shapes and appearances for drug release through a [111]
diffusion-controlled release mechanism
Intraocular inserts and I-vation: a titanium helical implant anchored within the sclera, containing drugs in the coating [152]
implants
Vitrasert (Retisert): a device sutured to the sclera with the drug reservoir inserted into the vitreous cavity [150,152]
Renexus (NT-501): a tube-shaped device with RRE cells to secrete CNTF to treat retinitis pigmentosa and dry [150,152]
AMD
Iluvien: a rod-shaped device for sustained delivery of fluocinolone acetonide from both ends of the device to [148,152]
treat diabetic macular edema
Capsule drug device: a refillable device with a ring shape inserted in the lens capsule to deliver Avastin [153]
Microneedles: stainless-steel microneedles inserted halfway into the cornea or scleral for 20 s to 2 min to [157,158]
dissolve drug-containing coatings
MEMS devices PDMS-based devices with a refillable drug reservoir, flexible cannula, check valve, and suture tabs to release [165–169]
drug by manual actuation or electrolysis pumps
Magnetically actuated PDMS devices composed of a drug-loaded microreservoir sealed by a thin elastic [173]
magnetic PDMS membrane with a laser-drilled microaperture for drug release
Oral delivery Bioadhesive intestinal Mucoadhesive disks with layered structure for sticking to the intestinal wall to prolonging drug release [176–178]
patch-like devices duration
Microfabricated PMMA devices containing single or multiple microdrug reservoirs [179–186]
Wireless endoscopic Swallowable capsule endoscopes usually consisting of a drug reservoir, a piston, and an energy [198–201]
capsules producer-transmission system for triggered drug release in the GI tract
Capsule endoscopes with a set of legs capable of active and controllable locomotion [202–205]
Capsule devices with magnetic parts to release drugs controlled by the external magnetic field [207–210]
Self-folding Stimuli-responsive self-folding microdevices with patterned rigid segments connecting with [86,87]
microcontainers flexible polymeric hinges
Transdermal Microneedles Four general types of microneedles: solid microneedles for skin pretreatment, drug-coated microneedles, [225–234]
delivery dissolvable microneedles, and hollow microneedles
Other designs of microneedles: biphasic microneedles with enhanced adhesion strength in the skin, [235–240]
bendable microneedles to avoid breakage and detachment, rapidly separating microneedles to optimize
dissolvable microneedles, hollow silicon microneedles with a flexible reservoir for manually controlled drug
delivery, a smart wearable microneedle system for diabetes monitoring and therapy
Optical lens-microneedle array for percutaneous light delivery for dermatologic applications and [241]
photodynamic skin cancer therapy
Other transdermal Stretchable patch-type devices with multiple reservoirs to release drug by bending and stretching the device [242]
devices to deform the reservoir
MEMS-based devices for electric fields triggered release with actuate shrinking of an electrosensitive [243]
hydrogel
Implantable Sustained delivery Microgeometry-based devices comprising of three functional layers and an array of micro-orifices to control [244]
devices devices drug diffusion
Silicon nanochannel membranes controlled drug release which is independent of the concentration [31,32,245]
gradient
Pulsatile delivery Drug layers separated by degradable blank polymer layers to achieve sequential and pulsatile drug release [246,247]
devices
Electrochemically activated and electrothermally activated microchips for pulsatile drug release [30,251]
Composite membranes embedded with thermosensitive nanogels and magnetic nanoparticles to control [96,254]
drug release by an oscillating magnetic field
Device with thermally responsive hydrogel for ultrasound triggered drug release [255]

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (11 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Table 1. Continued.

Administration Device categories Characteristics Ref.


routes
Magnetic fields controlled movement of Fe3O4 particles to block/open device pores for reversible switching [97]
of drug flux
MEMS devices with pump-like release mechanisms using magnetic films, magnetic blocks, magnetic [103,109,257]
sponges, or magnetic actuated plungers
Devices applied to Intracerebral delivery: microprobes with microfluidic channels for controlled drug infusion [264]
specific sites
Intracochlear delivery: a microfluidic reciprocating delivery device with a single microcannula to release drug [270]
to the cochlea
Intraoral delivery: a mechatronic device as part of a dental prosthesis for drug release through the buccal [271,272]
mucosa
Vaginal/intra- Vaginal rings Sandwich type rings consisting of a drug-containing central core surrounded by a drug-free outer layer of [279]
uterine delivery polymer
Vaginal rings with compartments for loading drugs in different forms such as silicone rods, compressed [280]
tablets, and lyophilized gels
A device with a flux controlled pump driven by swelling of drug-containing polymers [281]
Intrauterine devices Mirena system: a plastic device with a T-shaped polyethylene frame and drugs in the vertical part [285]
T-shaped devices with copper nanoparticles dispersed in plastic matrix
Other delivery Intranasal olfactory A system containing an aerosol generator (inhaler), a charging chamber, a focusing chamber, and external [286]
routes delivery systems electrodes to guide drugs to the targeted olfactory receptor
Surgery sutures A suture surrounded by a drug-containing PLGA sheet for sustained drug release without sacrificing the [287]
mechanical strength of the suture
Devices for rectal drug A hollow-type suppository made of oleaginous base with different gel agents to prolong the plasma [289]
delivery concentration of drug

a probe-type electrode, which has to be pressed on the eye offers a novel approach for treatment of other anterior condi-
manually. The drug release from these devices is through tions.[111] The potential advantages of using PPs over topical
a large area on the eyeball, raising a risk of damage to the drug delivery include reduction in loss of drug due to lacrimal
sensitive cornea or sclera. Recently, a simple device based on drainage of drug, enhanced efficacy, and enhanced patient
PDMS in a cup shape with a planar poly(3,4-ethylenedioxy- compliance.
thiophene) electrode was created by the microfabrication tech- PPs can be classified into two kinds: semipermanent and
nology (Figure 6A).[145] Due to its small size, the device can temporary, depending on the material used for their prepa-
be placed under the eyelid and deliver drugs into the vitreous ration. Semipermanent PPs are made from silicone, HEMA,
cavity through a small area of the sclera, reducing the possi- PCL, or Teflon and intended for six-month use. Removal of
bility of tissue damage. The in vivo drug delivery test showed such plugs is accomplished by either flushing or being man-
high iontophoretic efficiency of 396 ng mL−1 Mn2+, which ually removed by a physician. Temporary PPs that are made
was achieved compared to the delivery result (2.69 ng mL−1) from animal collagen last for 4–10 d and dissolve. To give drug
without iontophoresis.[146] delivery ability to PPs, a common method is to load drugs
within the core of the PPs coated with a layer of material that is
impermeable to the drug and tear fluid on all sides except the
3.1.3. Punctal Plugs head portion. The release of the drug from PPs is through a
diffusion-controlled release from the head cross-section facing
Punctal plugs (PPs), also called lacrimal plugs, are very tiny to the tears (Figure 4C(a)).[111] The forms of the drug can be
biocompatible devices in the treatment of dry eye syndrome solutions, suspensions, microemulsions, nanoparticles, or
since 1975.[111] They are used to block tear drainage through liposomes. For some plugs, the drug is loaded by soaking the
the canaliculi by inserting into the lid puncta, which have been plug in drug solution before insertion. However, the quantity
proved in the improvement of tear film stability and tear osmo- of loaded drug is limited due to the small surface area. In a
larity and have good acceptance by both patients and physi- report from Gupta and Chauhan,[147] a three-month zero-order
cians.[147] In addition to the physical occlusion of puncta, the release of cyclosporine A at a rate of 3 µg d−1 was achieved
administration of drugs such as cyclosporine A (an immune with a cylindrical plug consisting of HEMA core covered by an
suppressant) has shown promising results for the dry eyes. impermeable silicone shell. A mathematical model was also
Thus, the use of PPs for the simultaneous delivery of oph- built up to estimate of the drug release behaviors from the
thalmic medications to the tear fluid and the nasolacrimal duct PPs.[147]

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (12 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 6.  Different types of devices for ocular drug delivery. A) Schematic view of the ocular iontophoretic device that can be placed on a small area of the
eyeball, allowing ions penetration into the vitreous cavity by an electric field through the corneal epidermis. Reproduced with permission.[145] Copyright
2016, Elsevier. B) Selected drug delivery devices and their locations in the eye. Reproduced with permission.[153,158] Copyright 2010 and 2016, Elsevier.
C) MEMS ocular drug delivery pumps. (a) Illustration of an implanted passive MEMS pump.[165–167] Reproduced with permission.[167] Copyright 2009,
Springer; (b) Cross-section of an ocular drug delivery with the electrolysis pump.[168,169] Reproduced with permission.[169] Copyright 2010, Informa Health-
care; D) Conceptual illustration of a magnetically controlled MEMS device and its working principle. Reproduced with permission.[173] Copyright 2013, IEEE.

Although the advantages of PPs as ocular drug delivery newer and effective PPs with different shapes and appear-
devices have been demonstrated, some complications such ances have been fabricated to overcome some of the current
as epiphora, corneal abrasion, plus extrusion and suppu- drawbacks (Figure 4C(c)).[111] Some of them are under clin-
rative canaliculitis have also been observed. To date, many ical trials and some are already commercially available for

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (13 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

different purposes of application (tear retention or sustained non-biodegradable devices are anchored on the sclera and a
drug release). removal may be needed to implant other devices if required.
Iluvien is a rod-shaped device for sustained delivery of
fluocinolone acetonide to treat diabetic macular edema
3.1.4. Intraocular Inserts/Implants (Figure 6B).[108,148,152] The drug is encapsulated in a PVA matrix
inside a polyimide tube and then released from the membrane
In addition to the noninvasive means, some invasive routes caps on both ends of the device. Unlike the implant Retisert,
of drug administration have also been under investigation, Iluvien is a miniaturized injectable insert (3.5 mm in length
including implantable and injectable devices, designed to keep and 0.37 mm in diameter) which is small enough to be injected
effective therapeutic levels in eyes and reduce the frequency into the vitreous through a 25-gauge needle without any
of conventional intraocular injections. Using the sclera as an sutures. The drug release can last for up to 36 months with a
anchor site, various devices have been designed for intravit- rate of about 0.6 µg d−1. However, due to the non-biodegrada-
real release of different drugs to the posterior segment of the bility of Iluvien, the device would remain in the vitreous humor
eye.[148,149] after the drug payload is exhausted. The surgical removal of
I-vation is a titanium helical implant (0.5 mm long and this insert has not been reported so far. Different biodegrad-
0.21 mm wide) containing triamcinolone acetonide in the able devices have been designed and tested in order to over-
coating of nondegradable polymers (Figure 6B).[150,151] It can be come the issues associated with non-biodegradable implants.
implanted and anchored within the sclera through a 25-gauge Ozurdex is the first biodegradable intravitreal insert approved
needle stick. The helical structure of the device increases the by the US Food and Drug Administration.[66] It is a cylindrical
surface area available for drug coating and enables sutureless PLGA platform (6.5 mm in length and 0.45 mm in diameter)
operation of anchoring.[152] The implant was designed to elute containing 0.7 mg dexamethasone for treatment of macular
drug sustainedly for two years and showed its effectiveness edema and noninfectious uveitis (Figure 6B).[151,152] After injec-
in treating diabetic macular edema (DME) in phase 1 clinical tion of Ozurdex into the vitreous via a 22-gauge needle, the
trials. drug release is fast over the first two months and then becomes
Vitrasert is a marketed reservoir-type device to deliver slow in the following four months following the degradation of
ganciclovir for the treatment of cytomegalovirus retinitis PLGA. Although the drug release can be regulated by adjusting
(Figure 6B).[148,150,152] The drug pellet was encapsulated in a the ratio of lactide and glycolide in PLGA matrix, a zero-order
non-biodegradable polymer shell consisting of sandwiched release is still quite challenging. However, because the degra-
layers of polyvinyl alcohol (PVA) and ethylene vinyl acetate dation products of Ozurdex are water and carbon dioxide, no
(EVA). The PVA layer is permeable for drug diffusion and the surgical removal is required.
EVA layer is drug impermeable. The composite structure of two In addition to the intravitreal insert, an interesting drug
polymers allows the release of ganciclovir at a rate of 24 µg d−1 delivery device was designed and manufactured to insert in
for five to eight months. Retisert is another surgically implant- the lens capsule during cataract surgery for the treatment of
able device similar to Vitrasert, which is designed for the sus- wet AMD (Figure 6B).[153] The device consists of a ring-shaped
tained delivery of fluocinolone acetonide for the treatment of PMMA drug reservoir, a semipermeable membrane on one
chronic noninfectious posterior uveitis (Figure 6B).[148,150,152] side of the reservoir for controlled delivery, and silicone valves
It is 5 mm long, 2 mm wide, and 1.5 mm thick, and consists for drug refilling. The drug bevacizumab is dispersed within a
of a tiny tablet packed inside a silicone elastomer cup which PVA matrix and then placed in the reservoir. A near-zero order
is attached to a PVA suture tab. The silicone cup has an ori- release kinetic with a rate of 60–80 µg d−1 was achieved but only
fice for drug release and a PVA membrane between the tablet lasted for a few days. A long-term release profile is required to
and the orifice to avoid premature drug leakage. The drug is estimate the device feasibility for sustained drug release.[153]
released out slowly when the vitreous environment hydrates Mild invasive methods using miniaturized devices such
and dissolves the drug. The release rate is 0.3–0.4 µg d−1 for as microneedles have been proposed to better overcome the
≈30 months. Both Vitrasert and Retisert need to be sutured ocular barriers and enhance localization of the drug close to
to the sclera through a pars plana incision procedure with the the target tissues (e.g., retina).[154–156] For example, individual
drug reservoir inserted into the vitreous cavity. Renexus (also drug-coated microneedles with a length of 500–750 µm and a
known as NT-501) is a tube-shaped device containing geneti- width of 200 × 50 µm were developed for anterior and poste-
cally modified human retinal pigment epitheliums which can rior drug delivery through intracorneal and intrascleral routes
secrete recombinant human ciliary neurotrophic factor (CNTF) (Figure 6B).[157–159] The microneedles were fabricated by laser-
(Figure 6B).[150,152] The retinal pigment epithelium (RPE) cells cutting the needle structures from stainless-steel sheets and
are seeded in a polyethylene terephthalate scaffold surrounded then dip-coated in a drug/polymer solution. The drug-con-
by an outer shell made from a semipermeable polyether sulfone taining microneedles were inserted halfway into the cornea or
membrane. The device is implanted into the vitreous by a small sclera and then removed after 20 s to 2 min. The drug coating
sclera incision and fixed with a suture through a titanium loop could rapidly dissolve off the needles, generating a much higher
at one end of the device. The semipermeable membrane allows concentration in tissues compared to a topical administration of
outward diffusion of CNTF and protects the RPE cells from a free drug solution. And, the small abrasion formed at the site
host cellular immunologic attack. Clinical studies show that of microneedle insertion disappeared after 3 h. However, the
the device can release CNTF over 18 months for the treatment very low surface area of microneedles limits their drug carrying
of retinitis pigmentosa and dry AMD. The above-mentioned capacity. To address this limitation, microneedles with complex

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (14 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

through-thickness fenestrations (i.e., windows) were designed to milliamperes) allows a more precise drug release and makes
and fabricated using the titanium deep reactive ion etching tech- this device clinically practical for the treatment of chronic
nique. The fenestrated Ti MNs offered larger drug reservoirs and ocular diseases. Recently, a triboelectric nanogenerator (TENG)
consequently increased drug carrying capacity up to fivefold rela- device that can harvest ambient mechanical energy and transfer
tive to the solid counterpart.[157,158] In order to facilitate infusion it into electricity was introduced to offer the power for the elec-
of drug solutions such as nanoparticle and microparticle (20 nm trolysis pump, giving a self-powered ability and battery-less
to 10 µm) suspensions into the eyes, hollow glass and metal possibility to the current system. The concept of TENG-based
microneedles were developed and tested in ex vivo experiments self-powered device was demonstrated with the ex vivo trans-
on rabbit, pig, and human eyes.[160–163] For drug delivery, the sclera drug delivery in porcine eyes by generating the biokinetic
hollow microneedles were initially inserted into the sclera and energies of human hands.[170]
then partially retracted to enable drug solution infusion into the The prototype of the refillable MEMS-based device (the
tissue. An infused volume of 10–35 µL per microneedle could MicroPump system) has been realized by Replenish Inc. (Pasa-
be achieved, which was affected by the microneedle length, dena, CA) to deliver a programmable microdose of medicine
applied pressure, and particle size in the fluid. The infused solu- (such as ranibizumab) into the vitreous cavity for DME.[171,172]
tion using microneedles forms a drug depot within the sclera The surgical feasibility and safety of the device have been dem-
and gradually diffuses drug to neighboring tissues, such as onstrated in an animal model for up to one year and then in
choroid, retina, or ciliary body.[160,161] In general, the drug mole­ human patients with retinal disease for 90 d. As the Micro-
cules were primarily cleared from the suprachoroidal space in Pump device is supposed to be used for five years or longer
1 d, while the drugs encapsulated in particles could prolong the before replacement, long-term clinical studies are necessary
clearance period to one to two months. Microneedles have dem- to evaluate the stability, sustained function, and in-office drug
onstrated a minimally invasive means for localized ocular drug refilling of the device.
delivery. However, it was found that solution infusion by hollow Another MEMS-based ocular implant is a magnetically con-
microneedles caused more pain than intravitreal injection (to trolled MEMS device capable of on-demand release of an anti-
be discussed subsequently), because of distension by the drug proliferative drug, docetaxel to the posterior segment of the
solution. This may further limit the acceptable dose each time eye (Figure 6D).[173] The device is composed of a drug-loaded
and require repeated injections. Moreover, further research on microreservoir (6 mm in diameter and 580 µm in depth) sealed
optimization of microneedle designs, drug formulations, and by a 40 µm thick elastic magnetic PDMS membrane with a
operation procedures is needed to enable controlled and long- laser-drilled aperture (100 × 100 µm2) in its center as an outlet
term drug delivery. of drug release. By applying a magnetic field from the bottom
of the device, the PDMS membrane incorporated with iron
oxide nanoparticles deforms and pushes the drug solution out
3.1.5. MEMS-Based Devices from the reservoir. The amount of released drug is tunable by
changing the strength and duration of the applied magnetic
As noted above, a long-term sustained drug release is able to be field. Ex vivo release studies performed on human cadaveric
achieved with the currently available implants/inserts. Beyond this, eyes demonstrated a promising application of this device for the
benefitting from MEMS technologies, more sophisticated small- treatment of ocular posterior segment diseases such as diabetic
scale devices may now be fabricated for ocular drug delivery.[164] retinopathy by controlled drug delivery through an external
Meng and co-workers developed and tested a MEMS magnetic actuation. Likewise, the safety and reliability in the
device which was an assembled three-layer structure made long-term use of the device need to be further investigated.
from PDMS containing a refillable drug reservoir, flexi­ ble
cannula, check valve (a one way valve), and suture tabs
(Figure 6C(a)).[165–167] The device can be implanted underneath 3.2. Oral Delivery
the conjunctiva with the cannula outlet inserted through an
incision into either the anterior or posterior segment for dif- Oral drug delivery is a convenient and highly patient-compliant
ferent treatment requirements. The first generation of this route of drug administration among various conventional
device was designed for manually actuated drug release by delivery routes such as invasive injection manners (intravenous
pressing the reservoir with a finger. When the pressure was and intramuscular). However, a unique set of physiological bar-
high enough, the valve was opened to discharge drug solu- riers in the GI tract limit the overall efficacy of drugs delivered
tion. The drug refilling was accomplished by puncturing the through the oral route. The acidic environment of the stomach
reservoir layer using a 30-gauge needle attached to a syringe. combined with an array of intestinal enzymes may cause drug
Obviously, manual actuation could not ensure precise control degradation, especially for the active therapeutics such as pro-
over the amount of drug delivered. Later, an improvement tein and peptides. The adherent mucus layer and tight junc-
was made by incorporating an electrolysis pump and batteries tions of intestinal epithelium limit permeation of drugs with
into the device (Figure 6C(b)).[168,169] The pump can generate high molecular weights. Additionally, the shear forces and
hydrogen and oxygen gas through electrolysis of water, which flow conditions due to peristalsis of the GI tract may further
increases the internal pressure in the reservoir and thus decrease drug retention and result in low bioavailability. To
pushing drug out from the cannula. When the pump is turned overcome these obstacles, different delivery devices with special
off, these gases recombine to form water. The electrically con- design and precise engineering have been fabricated through
trolled manner with low power requirements (microamperes microfabrication for safe and effective oral drug delivery.[174,175]

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (15 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

3.2.1. Bioadhesive Intestinal Patch-like Devices but large enough to avoid endocytosis. The bioadhesion ability
of the device is achieved by conjugating bioadhesive tomato
Inspired by the success of transdermal patches, some similar lectin to the surface of reservoir side after chemical modifica-
mucoadhesive patch-like systems have been created for local tion. The drug is loaded in the device by forming hydrogel/drug
drug delivery in the GI lumen. These patch systems usually matrix in the reservoir via photolithography. By choosing a spe-
consist of several layers that have different functions including cific type of hydrogel material, the drug release behaviors from
adhesion, drug encapsulation, and protection from the sur- the device can be predetermined and tuned. When loading the
roundings. For example, a three-layer patch device was devel- different drugs with different hydrogels in the separated reser-
oped by Mitragotri and co-workers for oral protein delivery, voirs of a device, independent controlled release of each drug
which was prepared by sandwiching a layer of drug between a can be realized. The device was further improved by being
layer of a mucoadhesive polymer and a polymer layer with poor sealed with a nanostraw membrane with which enhanced adhe-
permeability (Figure 7A).[176–178] The mucoadhesive layer made sion of the device to the epithelial tissue and limited influx
of carbopol and pectin allows the device to stick to the intestinal of outside molecules into the device reservoir were observed
wall. The backing layer was made of ethyl cellulose (EC), which (Figure 7B(d)).[187] All of the features of the planar microdevice
is relatively impermeable compared to the mucoadhesive layer, delivery system, including bioadhesion ability, unidirectional
allowing the drug to be released primarily from the mucoadhe- controlled release, and decreased drug leakage, enable a local
sive side into the epithelia layer and preventing drug diffusion sustained drug release with enhanced therapeutic bioavail-
into the outer lumen. The backing layer, at the same time, may ability in the GI tract by oral route.
protect drug from enzymatic degradation. The patches with a
square or circle shape (2–4 mm2 large and 40 µm thick) could
be encapsulated in a capsule and delivered into the intestine 3.2.2. Wireless Endoscopic Capsules
(Figure 7A(b)).[177] The design of planar and layered structure
of the device offers several advantages over standard tablets or Swallowable wireless endoscopic capsules are types of small
mucoadhesive microspheres. Specifically, compared to micro- device for diagnostic inspection of the GI tract and in some
spheres with limited contact area, the patches provide larger cases, a drug chamber is incorporated in the device to per-
surface area due to the planar design, thereby enhancing their form a targeted therapeutic treatment.[188–190] Various commer-
adhesion ability on the intestinal wall and prolonging the dura- cial prototypes of capsule endoscope such as IntelliCap,[191,192]
tion of drug release at the target site. The flat design with InteliSite,[193] Pulsincap,[194] and Enterion capsule,[195] have been
minimized side area also decreases the shear disturbances designed and fabricated to achieve triggered drug release via
from the flow of liquids through the intestine. Unlike multi- different mechanisms. These devices generally consist of a cyl-
directional release from a spherical system, the layered struc- inder polymer capsule with an embedded assembly of a drug
ture of the patch with the low permeable backing film allows reservoir, a piston, and an energy producer-transmission system.
unidirectional release of drug toward the intestinal epithelium Drug release from the devices is triggered by the piston move-
and reduces loss of drug into the intestinal lumen. In vitro ment driven using self-produced energy or energy transformed
experiments with rat intestine showed that trans-lumenal flux from the external stimuli. For example, the InteliSite capsule
of model drugs from the patches was about 100-fold higher (10 mm in diameter and 35 mm in length) contains a drug res-
than that from a solution form. The potential drug leakage ervoir of 0.8 mL in volume adjoining a compressed spring that
from the side edges of the patch delivery system is one unre- is held with two shape memory alloy wire clips.[193] Once acti-
solved issue for the layered device. A simple solution would be vated by the remote antenna for 2 min, the wire clips deform
to coat individual patches with materials of low permeability, to let the spring propel the inner drug reservoir out and away
which was later achieved by Mitragotri and co-workers.[178] In from the capsule body, releasing contained drugs. Similarly, in
the new system, the drug was dispersed in a mucoadhesive other capsule endoscopes, a thermosensitive spring made of a
matrix made of carbopol, pectin, and sodium carboxymethylcel- shape memory alloy,[196] or a compressed stretchable compo-
lulose by pressing the mixture into a 400 µm thick disk. The nent[197] is used to drive the piston and push the drug out of the
disk was then coated on all sides but one with EC to form a reservoir. In addition to the spring-like mechanics that acts on
layer of about 50 µm. The protective coat minimized drug loss the piston, the gas pressure is also used to cause piston move-
and resulted in significant bioavailability enhancement in por- ment. One example was reported by Gröning et al., in which the
cine models. gas was produced by the electrolysis of hydrochloric acid and/
Similar to the patch device introduced above, another type or sulfuric acid in the capsule (Figure 7C(a)).[198,199] The energy
of bioadhesive intestinal device has been designed and investi- triggering the electrolysis was received from an oscillating cir-
gated by Desai and co-workers, which contains single[179–184] or cuit located inside the capsule through the resonance with an
multiple microdrug reservoirs[67,185,186] to incorporate a number external high frequency transmitter (24–27 MHz). Accordingly,
of drugs or biomolecules of interest (Figure 7B). The drug res- the drug release could get controlled. Another example was
ervoir is constructed mainly with PMMA through a series of from Pi et al.’s reports.[200,201] They developed a solid propellant
standard microfabrication process including photolithography, microthruster that can be ignited by an external radio frequency
deposition, and reactive ion etching. The size of the devices is signal to convert the solid propellant into gas, offering the pro-
designed on a microscale level and the shape can be tailored pulsion energy for the piston (Figure 7C(b)).[201] The produced
according different requirements, ensuring that the device is gas could provide sufficient pressure to empty the drug reser-
small enough to have adequate contact with the intestinal wall voir and decrease possible drug reflux.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (16 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 7.  Various devices for oral drug delivery. A) Layered intestinal patches.[176–178] (a) Schematic representation of the patch design; (b) Images of
capsules containing patches with the backing membrane is stained with sulforhodamine B, and a schematic representation of the drug delivery using
intestinal patches. Reproduced with permission.[177] Copyright 2004, Elsevier. B) Asymmetric microfabricated devices. (a) Schematic representation
of spherical particles and microdevice interface with intestinal epithelial cell surface. Reproduced with permission.[186] Copyright 2014, Wiley-VCH;
(b) Patches with a single reservoir[184] and (c) multiple reservoirs.[185] Reproduced with permission.[184,185] Copyright 2009 and 2012, Wiley-VCH; (d) An
asymmetric microdevice with nanostraw structure. Reproduced with permission.[187] Copyright 2016, American Chemical Society. C) Endoscopic cap-
sules for drug delivery in the gastrointestinal (GI) tract. (a) Schematic diagram of a remote-controlled capsule with a gas producing cell and a high
frequency receiver to control drug release. Reproduced with permission.[199] Copyright 2009, Elsevier; (b) Schematic diagram of a solid propellant micro-
thruster. Reproduced with permission.[201] Copyright 2010, Elsevier; Endoscopic capsules of (c) twelve legged design[205] and (d) two legged rounded
shaped design with a needle. Reproduced with permission.[204,206] Copyright 2009 and 2013, IEEE; (e) Capsule made of two magnets. Reproduced with
permission.[208] Copyright 2012, Elsevier; (f) Capsule with two internal on-board permanent magnets. Reproduced with permission.[209] Copyright 2012,
IEEE. D) Microcontainers: (a) Self-folding microcontainers. Reproduced with permission.[86] Copyright 2008, Royal Society of Chemistry; (b) Stimuli-
responsive theragrippers. Reproduced with permission.[87] Copyright 2014, Wiley-VCH.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (17 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Most existing capsule endoscopes move by the natural the volume of the drug chamber, decreasing drug leakage,
peristalsis of the GI tract, known as passive locomotion. This miniaturization of the device, and enhancing the repeatability
system has a lack of good controllability over their position, of drug release.
orientation, and moving speed. This limits their application
for advanced diagnostic and therapeutic purposes at the target
area in different sections of the GI tract. To overcome this 3.2.3. Self-Folding Microcontainers
problem, several systems capable of active and controllable
locomotion have been proposed. One of these systems uses a Microdevices with stimuli-responsive self-folding abilities are
legged mechanism with small brushless DC motors powered able to be used for encapsulation and delivery of drugs. Gra-
by an on-board battery.[202–204] A set of legs in the capsule can cias Laboratory has developed various polyhedral shaped,
be deployed to resist peristaltic force in the GI tract such as the self-folding microcontainers using a hybrid fabrication method-
intestine. The experiment in a porcine colon demonstrated that ology of photolithography and self-assembly.[211–214] The micro-
the capsule with four legs could travel a distance of 15 cm in containers consist of patterned rigid segments (metallic or
a period of 5 min, and the speed was able to be adjusted from polymeric) connecting with flexible polymeric hinges that are
10 to 40 mm min−1.[203] The four-leg device was later improved sensitive to temperature. Increasing the temperature of envi-
to achieve high propulsion by increasing the number of legs to ronment enables spontaneous transformation of 2D templates
12 (Figure 7C(c)).[205] In another device, besides an anchoring into 3D structures, which allows parallel loading of drugs or bio-
system, a needle positioning mechanism[204,206] was incorpo- logical objects. The untethered nature of these microcontainers
rated in the capsule for targeted dosing of 1 mL of medication allows free or remotely guided travel of the devices in the GI
to the GI tract (Figure 7C(d)). When the device is anchored tract. The encapsulated drugs can be released from the pores in
after its two round-shaped legs are fully deployed, the needle the porous walls of the microcontainer (Figure 7D(a)).[86] Photo-
positioning mechanism allows the needle to penetrate into the lithographic patterning enables precise control over the proper-
GI tract wall to inject medication from any one of 16 predefined ties of the microcontainers including the wall thickness, sizes,
outlet ports.[204] shapes, and porosity, all of which are important for controlled
The above capsule endoscopes to some extent achieve the drug delivery applications. A microcontainer referred to as a
controlled drug release in the GI tract. However, because all theragripper (Figure 7D(b)) was fabricated for drug delivery in
the components including on-board batteries and electronic/ the stomach of a pig after griping into tissue with its stiff tips
mechanical systems need to be integrated in a small cap- and the drug release could last for up to 7 d, which offers an
sule, there usually is limited volume left for drug loading. To exciting strategy to treat the various diseases of the intestine.[87]
enhance the capability of drug loading and maintain the locali-
zation functionality, a capsule device using purely magnetic
effects was proposed.[207,208] The capsule was comprised of two 3.3. Transdermal Delivery
metallic parts (Figure 7C(e)).[208] In magnetized state, the two
metallic parts were attached to each other and kept the capsule Skin is the largest accessible organ of the human body and
closed when travelling through the GI tract. The position of the offers a compliant interface for painless drug delivery by
capsule could be monitored. Once the capsule reached the area methods such as transdermal patches. Drug delivery across
of interest, it was opened and released drugs by applying an skin has a variety of advantages including avoidance of first-
external magnetic field to demagnetize the metallic parts. As pass metabolism and drug degradation of the GI tract com-
the device just consists of two magnetic capsule shells, it offers pared with the oral route, as well as less pain and reduced side
more volume as the drug reservoir compared to the device effects due to transiently high plasma drug concentration com-
with the same size. One drawback of this system is that drug pared with hypodermic injections.[215] In addition, transdermal
release is a one-time release event at one location. A design systems can provide sustained drug release for long periods of
that can give multiple doses in a controlled way may provide time and can be self-administered, highly improving patient
more possibilities in practical applications, such as allowing compliance. The greatest challenge for transdermal delivery
drug delivery to more areas in the GI tract by a single cap- might be that only small hydrophobic drugs (molecular weight
sule. A prototype of a magnetically actuated soft capsule was <500 Da) can effectively be delivered by this route because the
designed and investigated for this purpose.[209,210] The device resistance to drug transport resulted from the outermost layer
contains two permanent magnets in two ends of the capsule, of skin, known as the stratum corneum lipid bilayers.[216] Much
and the drug-containing chamber is situated between the effort has been made in order to overcome this barrier by devel-
two magnetic heads (Figure 7C(f)).[209] When the chamber is oping new technologies involving microfabricated transdermal
compressed by the movable magnetic head under an external devices.[217–219]
magnetic field, the drug is released through the slits of the
chamber. When the external magnetic field moves away, the
chamber can return to its previous shape and stop the drug 3.3.1. Microneedles
release by closing the slits. This process can be repeated until
the chamber is fully emptied. If the external magnetic field is Microneedles, usually an array of microscopic needles on a base
strong enough, the whole drug can also be released out due substrate, have been developed as a minimally invasive means
to the collapsing of the chamber. Despite the feasibility of this to deliver drugs into the skin. They are designed to be long
system, further improvement is still needed such as increasing enough to penetrate through the skin’s stratum corneum and

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (18 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

create holes for drugs permeation, but short enough to avoid with a stretchable base that contains four soft beam pillars
stimulating the nerves in the dermis, therefore achieving pain- made of PDMS.[236] These pillars may serve as anchors to fix
less and less bleeding drug delivery.[220,221] Earlier microneedles microneedles and allow bending of the microneedles when the
were made of silicon due to easy adaptability to microelectronic lateral force applied onto the microneedle exceeds the threshold,
fabrication process of silicon etching.[222] However, issues avoiding the breakage and detachment of the microneedle. This
associated with fragility of silicon microneedles arose. Subse- device may be applied on the joint such as elbow and knuckle
quently, metal materials such as titanium, nickel, and stain- for transdermal drug delivery in osteoporosis treatment. Rapi­dly
less steel were explored to decrease fracture of the tip of the separating microneedles (Figure 8B(c)) were fabricated to opti-
microneedle in operation. Fabrication techniques for such mize dissolvable microneedles, which usually need several min-
materials include 3D laser ablation, laser cutting, wet etching, utes for full dissolution in the skin tissue and are difficult to be
and metal electroplating, which are relatively expensive. Poly- completely inserted into the skin.[237,238] The rapidly separating
meric microneedles that can be fabricated using photolithog- microneedles are drug-incorporated PLA microneedles mounted
raphy and micromolding methods provide an alternative with on the top of solid microneedles.[238] Such stacked design not
low cost for large scale manufacturing.[222] only enabled full insertion of the drug-containing microneedles
In general, microneedles may be categorized into four into the skin due to the increased length, but also allowed rapid
different types including solid microneedles for skin separation of the top part from the bottom part to achieve the
pretreatment, drug-coated microneedles, dissolvable high drug delivery efficiency with a quick operation process
microneedles, and hollow microneedles (Figure 8A(a)).[223,224] compared to the counterpart. By bonding an array of hollow
Different designs of these microneedles enable drug silicon microneedles with a flexible PMDS reservoir, manually
delivery through different mechanisms. Solid microneedles controlled drug delivery was achieved by pressing the reservoir
(Figure 8A(b))[28,225,226] are used for pretreatment to enhance with a finger (Figure 8B(d)).[239] The pressure of a finger could
skin permeability of drugs by making microscopic holes in the push drug solution or microparticle suspension in the reservoir
skin. After piercing the skin and removing the micro­needles, into the skin through the hollow microneedles, giving a real
a drug formulation such as a transdermal patch or drug-con- time control on drug injection. Recently, Lee et al. reported a
taining gels may be applied to the skin, allowing diffusion smart wearable microneedle system for diabetes monitoring
of drug into the tissue through the holes. When an electric and therapy (Figure 8B(e)).[240] This system is an assembly of
field is applied, fast penetration of drugs can be realized by a heater, temperature, humidity, pH, and glucose sensors and
iontophoresis. Drug-coated microneedles (Figure 8A(c)) are drug-containing microneedles made of a phase-change mate-
solid microneedles containing a layer of drug coating, typi- rial. The sensors can monitor the sweat glucose concentration
cally a water-soluble formulation.[227–229] When inserting and trigger the embedded heaters to dissolve microneedles once
the microneedles in the skin, the drug coating starts to dis- the glucose concentration is high. As a result, the drug can be
solve and release drugs into the skin. After dissolution of the released from the bioresorbable microneedles in response to the
coating in a short time, the microneedles are removed. Dis- glucose sensing. Successful reduction in blood glucose level was
solvable microneedles (Figure 8A(d)) are a further improve- demonstrated by thermally actuated delivery of Metformin with
ment, in which the needle body is made of water-soluble or this smart system in diabetic mice.
biodegradable polymers with the drug of interest incorporated So far, microneedles for transdermal drug delivery have been
inside.[230,231] In this way, the microneedles could be left in investigated in vitro, in animal and in human. It has shown that
the skin after insertion and the encapsulated drug can slowly microneedle piercing can increase skin permeability by orders
be released along with the dissolution or degradation of the of magnitude for a broad range of drugs including small mole­
microneedles. As the whole microneedle body is used as a drug cular weight compounds, DNA, proteins, inactivated viruses,
reservoir, it greatly increases the drug amount in each dose. and nanoparticles. In addition to drug delivery, microneedles
The last type, hollow microneedles (Figure 8A(e)),[37,232–234] have been designed and fabricated for percutaneous light
can enable active drug fluid flow through the needle bore and delivery in the dermatologic applications of antimicrobial blue-
into the skin, which may achieve a fast drug delivery that can light therapy and photodynamic skin cancer therapy. The proto-
be easily modulated over time. type device, termed optical microneedle array (Figure 8C), con-
Some other designs were also proposed to improve the sisted of a press-molded PLA microneedle array bonded with
existing microneedle systems. For example, a biphasic a matched microlens array.[241] The microlens array was used
microneedle (Figure 8B(a)) was fabricated to enhance the to focus incident light into each microneedle at appropriate
adhesion strength between the microneedle patch and the converging angles to achieve an optimal intensity in the tissue.
skin.[235] The microneedle is composed of a poly(styrene)-block- The experiment with bovine tissues demonstrated that the
poly(acrylic acid) swellable tip and nonswellable polystyrene lens-assisted microneedles enabled a ninefold enhancement
core. After insertion of the microneedles in the skin, the needle of light delivery at the target depth compared to direct surface
tip swells to mechanically interlock with tissue and create illumination.
high adhesive strength, decreasing the premature detachment
of the device from the target site. Another design is aimed to
overcome the risk of microneedle breakage induced by buck- 3.3.2. Other Transdermal Devices
ling forces from the deformation or lateral movement of the
skin. To achieve this purpose, a bendable microneedle device There exist some stretchable patch-type release systems able to
(Figure 8B(b)) was made by fixing a rigid sharp microneedle be used in transdermal drug delivery. An example was reported

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (19 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 8.  Methods of drug delivery to the skin using microneedles (MNs). A) MNs of four basic designs for transdermal drug delivery. (a) Schematic diagram
of drug release mechanisms of different MNs; (b) Solid MNs. Reproduced with permission.[28,226] Copyright 1998, Wiley and Copyright 2004, Plenum Publishing
Corporation; (c) Coated MNs.[227–229] Reproduced with permission.[228,229] Copyright 2009 and 2010, Elsevier; (d) Dissolving MNs Reproduced with permis-
sion.[230,231] Copyright 2010, Nature Publishing Group and Copyright 2010, Springer; (e) Hollow MNs. Reproduced with permission.[37,234] Copyright 2006 and
2010, Springer. B) MNs with other special designs. (a) Swellable biphasic MNs with the mechanical interlocking mechanism. Reproduced with permission.[235]
Copyright 2013, Nature Publishing Group; (b) Bendable MNs. Reproduced with permission.[236] Copyright 2016, Wiley-VCH; (c) Rapidly separating MNs.
Reproduced with permission.[238] Copyright 2016, Elsevier; (d) A MN-based miniature syringe. Reproduced with permission.[239] Copyright 2009, Springer;
(e) Thermoresponsive MNs with a electrochemical device for diabetes monitoring and therapy. Reproduced with permission.[240] Copyright 2016, Nature Pub-
lishing Group. C) Optical lens-microneedle array for percutaneous light delivery. Reproduced with permission.[241] Copyright 2016, Optical Society of America.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (20 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

by Kim et al., in which mechanical strain from human body concentration follows Fick’s diffusion laws. The diffusion flux
motions was used as means of stimulation for drug release.[242] is proportional to the concentration gradient. However, when
The patch-type device is based on a stretchable layer with mul- the solute passes through pores or channels with the size com-
tiple reservoirs which can deform and undergo a decrease in parable to the molecular hydrodynamic radius of the solute,
volume by bending and stretching the device to release drug unexpected effects may occur and result in significant devia-
from the micro-orifices. The use of elastic microchannels with tions from the kinetics predicted by Fick’s laws. For example,
different channel lengths attached to the opening of the res- in the case of single file diffusion, the molecular flux is inde-
ervoir enables reduction of the diffusional leakage and better pendent of the concentration gradient, because the solute
control over the drug release. Moreover, the reservoirs of the molecules cannot pass each other in pores that approximate
device can be refilled with a microsyringe to achieve long-term the dimensions of the molecule itself. Utilizing this phenom-
drug release. Liu et al. presented a MEMS-based drug delivery enon, silicon nanochannel membranes (NMs) (Figure 9A(a))
device consisting of a metallic contact array with a meander have been created with top-down microfabrication methods to
structure.[243] This device is able to create a uniform electric control drug diffusion, which consist of arrays of parallel rec-
field to actuate shrinking of the electrosensitive polymethacrylic tangular channels with the smallest aspect ranging from 2 to
acid hydrogel, leading to drug release from the hydrogel matrix. 50 nm.[31,32,245] The NMs have been demonstrated to deliver
By controlling the applied voltage and its duration, the amount a wide range of therapeutic agents in a constant, controllable
of released drug may be controlled. way. As shown in Figure 9A(b), a discoidal implantable cap-
sule (260 µL drug reservoir, diameter = 15 mm, thickness =
4.5 mm) and a cylindrical one (1 mL drug reservoir, diameter =
3.4. Implantable Devices 11 mm, length = 26 mm), were fabricated for the constant and
sustained delivery of therapeutically relevant doses of different
Implantable drug delivery devices are designed to be placed in drugs in three animal models (mice, rats, and dogs) for up to
desired bodily compartments (for example, close to the target 70 d.[245] By tailoring the geometrical parameters and surface
region beneath the skin) and offer a prolonged drug effect by properties of the nanochannel according to the size and mole­
a sustained, preprogramed, or externally controlled way. This cular properties of each therapeutic agent, a constant release
approach to drug delivery is very appealing for many kinds of with appropriate release rates including zero-order release
drugs, particularly those which cannot be given via conven- could be obtained. And, the drug could be loaded in both liquid
tional routes and for patients with chronic diseases who need and solid form. Such NM implants represent a viable nanotech-
long-term and repeated dosing. nological approach for the controlled administration of various
drugs to improve the therapeutic efficacy of treatment.

3.4.1. Sustained Delivery Devices


3.4.2. Pulsatile Delivery Devices
Sustained long-term drug therapy is difficult to achieve by con-
ventional means such as daily oral dosing. Implantable devices, In numerous cases, sustained release is not an optimal method
however, offer a possible solution to this challenge. For most for drug administration. Instead, pulsatile release where drugs
local drug delivery systems made of biodegradable polymers, are delivered at variable time intervals, may work better in cer-
drug release relies on the degradation of the scaffold and usu- tain cases such as the treatment of diabetes via pulsatile insulin
ally exhibits burst release due to the high initial concentration. delivery, and alleviating pains according the patients’ needs
Such degradation controlled release is often limited by the in the therapy of chronic illness. A simple method of making
property of the scaffold polymer. However, through different devices with sequential and pulsatile delivery abilities is to
designs, devices made of the same material could provide an sequentially load drugs in a reservoir with a separating polymer
alternative measure of controlling drug release. An example is layer between two drug layers (Figure 9B(a)).[246,247] With the
a microgeometry-based drug delivery device reported by Ryu et degradation/erosion of the blank polymer layer, the drug may
al.[244] In their design, the device comprises of three functional be released sequentially in a pulsatile manner. The drug release
layers: a drug reservoir layer, a diffusion layer, and a diffusion timing can be programed by choosing polymers with different
control layer. The diffusion control layer contains an array of degradation speeds or varying the thickness of the polymer
micro-orifices through which the drug can diffuse out. Drug layer. This device has limitations that the drug release is basi-
release from the patch-type device could be modulated by the cally through a passive manner at predetermined release timing
variation of the microgeometries in the release control layer which cannot allow an active control over the release behaviors
such as the size, number, and spatial distribution of the micro- after administration.
orifices or the reservoirs. As it is made from biodegradable An alternative method for achieving pulsatile release is
PLGA, the device can release drugs in predetermined rates and to develop active devices which can be triggered by external
degrade away afterward finally after being implanted close to stimuli to release drug. One prototype of this kind of devices
the target area. (Figure 9B(b)) was designed and fabricated by Langer and co-
Similarly, subtly designed silicon nanochannel membranes workers.[30,91,248] It is an electrochemically activated microchip
have been fabricated to control diffusion kinetics of different comprised of an array of reservoirs in a silicon substrate. The
drugs. In general, diffusion of solutes in unconstrained cases outlet for drug release of each reservoir is sealed by a thin
from a region of higher concentration to a region of lower gold membrane that serves as an anode in an electrochemical

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (21 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 9.  A) Silicon nanochannel membranes. (a) Schematic diagram and the scanning electron microscope (SEM) image of membrane microstructure
and the diffusion path of molecules across the membrane. Reproduced with permission.[32] Copyright 2011, Springer; (b) Photograph and schematic
rendering of the discoidal (left) and cylindrical (right) implantable capsules with nanochannel membranes.[31,32,245] Reproduced with permission.[245]
Copyright 2013, Elsevier. B) Different devices for pulsatile drug release: (a) Multilayered devices for sequential drug release. Reproduced with permis-
sion.[247] Copyright 2015, Wiley-VCH; (b) Multiwell silicon-based drug-release device for electrochemically actuated drug delivery. Reproduced with
permission.[30] Copyright 1999, Nature Publishing Group; (c) The microchip-based drug delivery device (MicroCHIPS). Reproduced with permission.[250]
Copyright 2006, Nature Publishing Group. C) Actuated devices for pulsatile drug delivery: (a) Magnetically triggered nanocomposite membranes with
thermoresponsive nanogels and superparamagnetic nanoparticles. Reproduced with permission.[254] Copyright 2011, American Chemical Society;
(b) Magnetically triggered device for reversible controlled drug delivery. Reproduced with permission.[97] Copyright 2009, Wiley-VCH; (c) Ultrasound-
based approach for triggering implanted devices: cross-section diagram (top) and top-down photograph (bottom). Reproduced with permission.[255]
Copyright 2016, Nature Publishing Group.

triggering reaction. When an electric potential is applied to the current within microseconds, which is much faster than the
gold membrane in a solution containing physiological levels of electrochemical method. The implantable microchip-based
saline, the anode membrane dissolves away and releases the drug delivery device developed by MicroCHIPS (Figure 9B(c))
encapsulated drugs which could be loaded in solid, liquid, or gel has been demonstrated to have an excellent in vitro to in vivo
form. The release mechanism of this device allows the precisely release rate correlation.[251,252] It has also been tested in human
timed release of drugs from an implant. This prototype was fur- by being implanted subcutaneously in the abdomen to deliver
ther improved by using the electrothermal method to open res- pulsatile parathyroid hormone for osteoporosis treatment.[253]
ervoirs instead of electrochemical dissolution approach.[95,249,250] Some other devices were designed for the pulsatile delivery
The metal membrane (gold or a platinum and titanium lami- of therapeutic agents without the integration of a microbat-
nate) can be removed by resistive heating from an applied tery, multiplexing circuitry, and memory into the device. One

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (22 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 10.  A) Magnetically triggerable microspouter with a magnetic sponge for on-demand drug delivery. Reproduced with permission.[257] Copyright
2017, Wiley-VCH. B) Intracerebral drug delivery system (the NeuroMedicator) with microprobes. Reproduced with permission.[264] Copyright 2012,
Springer. C) Microfluidic reciprocating intracochlear drug delivery system with a flow pump and a miniature electromechanical actuator. Reproduced
with permission.[270] Copyright 2014, Royal Society of Chemistry. D) Exploded view of an intraoral drug delivery device and its prototype (“IntelliDrug”)
embedded in a partial lower jaw denture.[271,272] Reproduced with permission.[272] Copyright 2009, Springer.

type of such device was based on composite membranes made pump-like release mechanisms allow a quicker release by
of an ethylcellulose film with embedded thermosensitive pumping drug solutions out in a short time. By incorporating
poly(n-isopropyl acrylamide) nanogels and magnetic nanopar- magnetic materials with soft PDMS membranes, Chiao and co-
ticles reported by Hoare et al. (Figure 9C(a)).[96,254] When the workers have developed various microdevices for on-demand
membrane was heated by the embedded magnetic nanoparti- release of drugs with remotely controlled magnetic fields. In the
cles in the presence of an oscillating magnetic field, the nano- button-like device (Figure 3D),[104,105] PDMS was incorporated
gels shrank, increasing the permeability of the membrane for with magnetic microparticles, forming a magnetic membrane
drug release from the reservoir. A thermally responsive hydrogel to seal the drug-containing reservoir. When the membrane
(N-isopropylacrylamide-co-acrylamide (NiPAAm-co-AAm)) was deflected under external magnetic fields, the drug was released
also used for stimuli-controlled drug release. The hydrogel through a microaperture at the center of the membrane. In
was loaded with drugs and placed in a capsule. When heating the cylindrical device (Figure 4B),[109] a piece of rectangular
the capsule above body temperature with a focused ultrasound magnetic block was bonded at the center of PDMS mem-
transducer, the hydrogel contracted and released drugs through brane facing the reservoir, which created larger deformations
the opening of the capsule (Figure 9C(c)).[255] In another device, of the membrane under the same magnetic field compared
Fe3O4 particles are used to block/open the pores of a membrane to the device with the magnetic membrane. In their recent
covering on the reservoir by controlling their movement inside reports, magnetic PDMS sponges made of porous PDMS scaf-
the drug reservoir, achieving a reversible switching of drug flux folds with embedded magnetic microparticles were fabricated
and finally pulsatile release (Figure 9C(b)).[97] and used for remotely triggered drug release.[256,257] A power-
Drug release from the above devices is through a diffu- less drug delivery device termed a microspouter was prepared
sion manner after the device outlet is opened remotely, while to provide active and precise control of localized drug delivery

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (23 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

on demand (Figure 10A).[257] The microspouter consisted of a will switch the microvalve to allow drug release to the buccal
magnetic sponge to provide the force for drug release based on side. This device is convenient to be used, worn, removed,
magnetic field-induced reversible deformation, a reservoir for refilled, or reinserted in the oral cavity without the need for
the sponge installation and drug loading, and a soft membrane surgery. It can also be fixed to a mouth guard, which has been
for sealing the device. Following application of a magnetic field used to deliver naltrexone for the treatment of chronic diseases
to the microspouter, the shrinking of the sponge could trigger a and addiction.[273]
spouting of drug through a membrane’s microaperture and the
spouted dosage was highly repeatable and adjustable by varying
the number of magnetic actuations or the magnetic strength. 3.5. Vaginal/Intrauterine Delivery
Controlled release was demonstrated in experiments in vitro
and ex vivo. Other research describing magnetic pump for on- Site specific delivery to the female reproductive tract (vaginal
demand drug administration was reported by Lee et al.[103] In or intrauterine drug delivery) has been studied not only for
their design, a magnetic actuator equipped with a plunger and contraceptive purposes but also for treatment of diseases and
a barrel was employed to pump insulin solution out the device disorders.[274,275] In addition to localized drug delivery for
through the to-and-fro movement of the actuator under an antifungal or contraceptive purpose, the vaginal route is suit-
external magnetic field. The implantable device was designed able for systemic administration of drugs due to its dense
to be refilled, which may prolong the service life of the device. vasculature and avoidance of first-pass metabolism and enzy-
matic degradation of drugs associated with the oral route.[276]
Vaginal rings are flexible, circular ring-shaped devices made
3.4.3. Devices Applied to Specific Sites of silicone elastomer or thermoplastic polymers for controlled
drug delivery after insertion in the vagina.[277,278] Compared to
Intracerebral Delivery: Poor permeability of high mass and polar common intravaginal drug formulations such as tablets, gels,
molecules across brain barriers is a major limiting factor for creams, and suppositories, vaginal rings allow sustained and
the pharmacological treatment of many brain disorders. Some long-term delivery of pharmaceutical substances, which can be
microprobes mostly combined with microelectrodes have been controlled by the user and does not interfere with any sexual
created to deliver bioactive compounds directly into the brain experiences. In simple vaginal rings, drug is homogeneously
or central nervous system and simultaneously record and dispersed within the circular ring polymeric matrix that gener-
manipulate neuronal activity.[258–263] Microfluidic channels are ally is 55 mm in diameter and 4–9 mm in cross-sectional diam-
fabricated within these probes, which can serve as drug depots eter. Such rings sometimes have an initial burst release due to
and pathways for drug discharge (Figure 10B).[264] Drug release the fast dissolution and release of drugs at the ring’s surface.
from such devices can be through ways of passive diffusion and To obtain stable drug release from vaginal rings, devices of new
controlled infusion. designs such as sandwich and reservoir type rings have been
Intracochlear Delivery: Direct delivery of drugs into the inner developed. The sandwich type rings consist of a drug-loaded
ear is required for many therapies.[265] However, the small polymer layer positioned between a nonmedicated inner central
space and limited fluid volume within the cochlea present core and a nonmedicated outer shell (Figure 11A).[279] The res-
a challenge for conventional drug delivery systems.[266,267] A ervoir design is a drug-containing central core surrounded by a
microfluidic reciprocating drug delivery device has been fabri- drug-free outer layer of polymer. The drug-containing reservoir
cated and tested to overcome this challenge, with which soluble in a single ring could either be an integrated core or several
drugs can be delivered to the cochlea via a single microcan- discontinuous segments for different drugs (Figure 11A(b)),
nula (Figure 10C).[268–270] The microcannula is connected to a thereby allowing delivery of multiple drugs from one device.[279]
closed microfluidic reciprocating circuit which allows infusing For example, in the report by Chen et al., hydroxychloroquine,
and withdrawing a constant volume of drug in a cyclic fashion. an anti-inflammatory and anti human immunodeficiency virus
This drug delivery strategy not only ensures in zero net volume (HIV) drug, was loaded in one or three reservoir segments in a
transfer to protect the delicate structures in ear from damaging vaginal ring to achieve near-zero-order release kinetics for the
changes in perilymph volume and pressure, but also achieves prevention of HIV transmission and other sexually transmitted
effective drug delivery through the diffusion and mixing of the infections.[279] Drug release rates from the sandwich or reser-
high concentrated drug with perilymph. The effective drug pen- voir type rings may be modified by changing the core diameter
etration of this device has been demonstrated with in vitro and or thickness of the nonmedicated coating.
in vivo studies in guinea pigs.[270] Vaginal rings have proven quite successful in delivery of
Intraoral Delivery: Dental delivery systems today are used for small lipophilic drugs such as steroids and spermicidal agents
the local treatment of diseases affecting the oral cavity itself which are not thermolabile because the manufacturing pro-
(e.g., periodontitis or fungal infections), or for systemic drug cess of vaginal rings usually involves temperatures ranging
delivery through the buccal mucosa. A mechatronic device (the 80–120 °C. However, it is challenging for conventional vaginal
IntelliDrug device) (Figure 10D) has been designed to be part rings to deliver macromolecular actives and hydrophilic drugs
of a dental prosthesis for intraoral drug delivery.[271,272] It is an due to poor solubility and diffusivity of these drugs in the matrix
assembly of a drug reservoir, an electronic control system, sen- polymers of vaginal rings. An insert vaginal ring was designed
sors, power supply, microvalves, and an osmotic pump. The and fabricated to address this issue, which contained some
water from the saliva enters the system through an osmotic compartments in the ring body for placing various drug-loaded
membrane, generating a pressure in the drug reservoir which inserts. As shown in Figure 11B, the drug-loaded inserts can

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (24 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 11.  Vaginal/intrauterine delivery devices. A) Schematic illustrating large- and small-matrix intravaginal rings with drug (orange) dispersed
within the polyether urethane matrix.[279] B) Silicone insert vaginal ring with three equidistant holes for loading drugs (left to right: silicone insert,
directly compressed tablet insert, and lyophilized insert). Reproduced with permission.[280] Copyright 2011, Elsevier. C) Intravaginal flux controlled
pump for drug release through polymer hydration and swelling. Reproduced with permission.[281] Copyright 2014, Springer. D) A schematic image of
T-shaped intrauterine device (Mirena system) for birth control. E) A copper-containing intrauterine device. Reproduced with permission.[285] Copyright
2011, Springer.

be silicone rods, compressed tablets with polymeric excipients, orifices, in which a drug-containing polymer pellet is placed.
and lyophilized gels.[280] The study demonstrated that both the The orifices on the chamber allow influx of water/vaginal fluid
release rate and duration of the model protein bovine serum to hydrate the pellet and efflux of drugs with the swollen poly-
albumin (BSA) could be tailored by choosing the insert type and mers. By altering the property of the swelling poly­mer and ori-
number. In particular, the lyophilized gel inserts are a proper fice diameter, drug release behaviors can be controlled. An oval
matrix for biological agents loading because of the mild manu- shaped vaginal retainer was used with this FCP to achieve a 30 d
facturing conditions. In another design, a flux controlled pump sustained release of insulin in female rabbits.
(FCP) was suggested (Figure 11C).[281] The FCP is composed Intrauterine devices are another type of topical drug delivery
of a cylinder chamber made of a rigid thermoplastic and two systems for women. The geometry of these devices basically

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (25 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

is designed as an inverted triangle shape to match the uterine


cavity. A commercial product is the Mirena system (Figure 11D),
a small, flexible, plastic device with a T-shaped polyethylene
frame, for sustained releasing of a progestin hormone (lev-
onorgestrel) into the uterus.[282] The vertical part of the device
is a reservoir consisting of a mixture of the drug and PDMS
surrounded by a silicone membrane for controlling drug
release. This device is able to slowly release levonorgestrel for
contraception or treatment of heavy menstrual bleeding for
up to five years. Copper-bearing intrauterine devices are also
widely used for the contraceptive purpose, which can release
cupric ions in uterine secretions to lead the spermatozoa to
lose activities.[283,284] The existing copper-bearing devices are
made by enwinding copper wires on the device body, which
usually causes side effects such as pain and bleeding due to
the fragmentation or rough surface of copper after long-term
use. Incorporating copper nanoparticles in plastic matrix of the
device may achieve controlled release of cupric ions and elimi-
nate the above-mentioned adverse factors (Figure 11E).[285]
Moreover, a custom-made fabrication of such drug-containing
matrix-type devices could be realized with 3D printing
technology.

3.6. Other Delivery Routes and Devices

To bypass the blood–brain–barrier and directly deliver medica-


tion to the brain and spinal cord, a new protocol was proposed
to efficiently deliver drugs by guiding electrophoretic drug par-
ticles to the olfactory region where the central nervous system
is in direct contact with the outer environment. The intranasal
olfactory delivery system is shown in Figure 12A.[286] It contains
an aerosol generator (inhaler) to form small pharmaceutical
particles, a charging chamber to make the particles acquire
a given number of charges, a focusing chamber to allow the
charged particles to form a finely focused beam, and finally
external electrodes to offer the electrophoretic force guiding the
particles to the targeted olfactory receptor. The feasibility of this
method was numerically evaluated in 2D and 3D nose models.
Figure 12.  A) An electrophoretic olfactory drug delivery system consisting
The study results indicated that olfactory drug delivery could be of components with four functions: aerosol generation (inhaler), particle
significantly enhanced after appropriate electrophoretic guid- charging, particle focusing, and particle navigation control. Reproduced
ance of the motion of drug particles. with permission.[286] Copyright 2014, Public Library of Science. B) Schematic
Surgery suture can be used as a drug delivery device to treat procedure for preparation of the drug delivery sutures and the SEM images
postoperative pain originating from the wound and avoid the of the suture. Reproduced with permission.[287] Copyright 2013, Elsevier.
side effects from taking drugs via the oral route or injection.
However, drug incorporation for the suture by solution dip-
ping, drug grafting, or encasing drugs directly in the suture especially for children and the elderly.[288] A hollow-type sup-
thread itself, somehow decreases the mechanical strength of pository was developed by Watanabe and co-workers to solve
the suture needed for wound closure. To address this problem, some drawbacks of conventional solid suppositories, such as
Lee et al. fabricated a drug delivery suture by physically assem- poor dose-volume control and undesired interaction of the
bling a PLGA sheet which contains pain-relief drugs onto a drug with base materials during preparation.[289] The shell of
clinically used surgical suture (Figure 12B).[287] This resulting the hollow-type suppository was made of oleaginous base alone
suture was able to achieve sustained drug release for effective or with gel agents of sodium alginate (Alg-Na), sodium poly-
pain relief without sacrificing the mechanical strength of the acrylate (PANa), or polyacrylate–PANa copolymer (PA–PANa).
suture. The drug release period could be varied for up to 6 d by The suppository could contain the drug (aminophylline) only
modifying the drug-loaded polymer sheet. or the drug with Alg-Na or PANa in the cavity (hollow space).
The rectal route is considered a good alternative to the oral The hollow-type suppository provided a prolonged plasma
route for certain groups of patients with difficulties of dys- concentration of drug in rabbits and could be used for rectal
phagia such as vomiting, nausea, or unconscious condition, administration of various drugs.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (26 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

4. Conclusions and Prospects Keywords


controlled release, design, drug delivery devices, MEMS, microfabrication
Drug release properties from many conventional delivery sys-
tems are currently unsatisfactory or at least, suboptimal, due Received: June 30, 2017
to poor drug biodistribution, unfavorable pharmacokinetics, Revised: August 2, 2017
systemic toxicity, and low therapeutic efficacy. This may arise Published online: October 9, 2017
from poor design, limited selectivity of materials for drug
scaffolds, weak control mechanism of drug release, or rela-
tively antiquated matrix/device fabrication methods. The fast [1] H. Rosen, T. Abribat, Nat. Rev. Drug Discovery 2005, 4, 381.
development of microfabrication technology such as MEMS [2] S. Sharma, A. J. Nijdam, P. M. Sinha, R. J. Walczak, X. Liu,
technology has speeded up the rise and evolution of innovative M. M.-C. Cheng, M. Ferrari, Expert Opin. Drug Delivery 2006, 3, 379.
drug delivery systems which have great potential to overcome [3] A. S. Hoffman, J. Controlled Release 2008, 132, 153.
these existing hurdles.[18,290] [4] Y. Yun, B. K. Lee, K. Park, Front. Chem. Sci. Eng. 2014, 8, 276.
Microfabrication technologies allow for greater miniaturiza- [5] J. West, Adv. Drug Delivery Rev. 2002, 54, 1225.
[6] C. P. McCoy, C. Brady, J. F. Cowley, S. M. McGlinchey,
tion of active delivery components such as valves and pumps
N. McGoldrick, D. J. Kinnear, G. P. Andrews, D. S. Jones, Expert
and construction of finer structures in a single device, which Opin. Drug Delivery 2010, 7, 605.
may not only reduce the volume and power required for a [7] A. N. Zelikin, C. Ehrhardt, A. M. Healy, Nat. Chem. 2016, 8, 997.
portable or implantable device but also introduce new control [8] M. W. Tibbitt, J. E. Dahlman, R. Langer, J. Am. Chem. Soc. 2016,
mechanisms for accurate dosing, complex release patterns, 138, 704.
and improved drug availability. A variety of microfabricated [9] H. Zhang, G. Wang, H. Yang, Expert Opin. Drug Delivery 2011, 8,
devices have been designed and fabricated over the years and 171.
demonstrated to be powerful tools for controlled delivery of [10] H. Zhang, R. Hao, X. Ren, L. Yu, H. Yang, H. Yu, RSC Adv. 2013,
drugs locally or systemically. For example, microfabricated 3, 22927.
microneedles have changed traditional drug delivery by trans- [11] Y. Brudno, D. J. Mooney, J. Controlled Release 2015, 219, 8.
[12] A. Santos, M. Sinn Aw, M. Bariana, T. Kumeria, Y. Wang, D. Losic,
dermal routes, offering painless delivery of macromolecules
J. Mater. Chem. B 2014, 2, 6157.
such as proteins, to cells, targeted regions, and systemically. [13] H. Zhang, C. Zhao, H. Cao, G. Wang, L. Song, G. Niu, H. Yang,
In addition, oral drug delivery microdevices fabricated using J. Ma, S. Zhu, Biomaterials 2010, 31, 5445.
novel MEMS and NEMS technology have exhibited poten- [14] C. Alvarez-Lorenzo, A. Concheiro, Chem. Commun. 2014, 50, 7743.
tial advantages such as patient compliance, rapid availability, [15] D. A. Lavan, D. M. Lynn, R. Langer, Discovery 2002, 1, 4987.
and low cost over the conventional pills.[18] The implantable [16] D. A. LaVan, T. McGuire, R. Langer, Nat. Biotechnol. 2003, 21,
microdevices with MEMS elements have shown promise in 1184.
sustained and on-demand drug release within the therapeutic [17] S. L. Tao, T. A. Desai, Adv. Drug Delivery Rev. 2003, 55, 315.
window through an external control manner. There devices pro- [18] M. Staples, K. Daniel, M. J. Cima, R. Langer, Pharm. Res. 2006, 23,
vide alternative routes of drug delivery that can benefit a large 847.
[19] R. S. Shawgo, A. C. R. Grayson, Y. Li, M. J. Cima, Curr. Opin. Solid
number of patients.
State Mater. Sci. 2002, 6, 329.
One of the long-term goals for drug delivery devices is to [20] E. Nuxoll, Adv. Drug Delivery Rev. 2013, 65, 1611.
integrate the controlled drug release functionality with sensor [21] J. Z. Hilt, N. A. Peppas, Int. J. Pharm. 2005, 306, 15.
technology, which can monitor continuously to physiological [22] K. M. Ainslie, T. A. Desai, Lab Chip 2008, 8, 1864.
changes and deliver precise amounts of therapeutic com- [23] G. Kotzar, M. Freas, P. Abel, A. Fleischman, S. Roy, C. Zorman,
pounds when needed, forming a closed loop of diagnosis and J. M. Moran, J. Melzak, Biomaterials 2002, 23, 2737.
therapy.[100,290] This may permit the personalized or custom- [24] G. Voskerician, M. S. Shive, R. S. Shawgo, H. Von Recum,
ized medicine to optimize treatment of patients. Additionally, J. M. Anderson, M. J. Cima, R. Langer, Biomaterials 2003, 24, 1959.
to produce viable devices for clinical applications, certain issues [25] L. Muthusubramaniam, R. Lowe, W. H. Fissell, L. Li,
such as the biocompatibility, long-term stability of performance, R. E. Marchant, T. A. Desai, S. Roy, Ann. Biomed. Eng. 2011, 39,
1296.
and replacement procedures, are especially important for new
[26] K. M. Ainslie, S. L. Tao, K. C. Popat, T. A. Desai, ACS Nano 2008,
devices, especially the implantable ones where tissue adhesion 2, 1076.
or polymeric degradation products may cause inflammation. [27] K. B. Vinayakumar, G. M. Hegde, M. M. Nayak, N. S. Dinesh,
Therefore, careful material selection and more in vivo assess- K. Rajanna, Microelectron. Eng. 2014, 128, 12.
ments are certainly needed when developing a new delivery [28] S. Henry, D. V. McAllister, M. G. Allen, M. R. Prausnitz, J. Pharm.
device in future. Sci. 1998, 87, 922.
However, microfabrication technologies with incomparable [29] A. J. Chung, Y. S. Huh, D. Erickson, Biomed. Microdevices 2009, 11,
advantages over other fabrication technologies are assured to 861.
significantly influence the future of drug delivery. This may [30] J. T. Santini Jr., M. J. Cima, R. Langer, Nature 1999, 397, 335.
have a considerable impact on next generation of life-saving/life- [31] F. Martin, R. Walczak, A. Boiarski, M. Cohen, T. West,
C. Cosentino, M. Ferrari, J. Controlled Release 2005, 102, 123.
enhancing medical therapies and the way people live their lives.
[32] A. Grattoni, H. Shen, D. Fine, A. Ziemys, J. S. Gill, L. Hudson,
S. Hosali, R. Goodall, X. Liu, M. Ferrari, Pharm. Res. 2011, 28,
292.
Conflict of Interest [33] N. F. Sheppard, D. J. Mears, S. W. Straka, J. Controlled Release
The authors declare no conflict of interest. 1996, 42, 15.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (27 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

[34] C. Iliescu, H. Taylor, M. Avram, J. Miao, S. Franssila, Biomicroflu- [67] S. L. Tao, M. W. Lubeley, T. A. Desai, J. Controlled Release 2003, 88,
idics 2012, 6, 16505. 215.
[35] P. F. O’Neill, A. Ben Azouz, M. Vázquez, J. Liu, S. Marczak, [68] A. C. R. Grayson, I. S. Choi, B. M. Tyler, P. P. Wang, H. Brem,
Z. Slouka, H. C. Chang, D. Diamond, D. Brabazon, Biomicrofluidics M. J. Cima, R. Langer, Nat. Mater. 2003, 2, 767.
2014, 8, 52112. [69] J. M. Seitz, M. Durisin, J. Goldman, J. W. Drelich, Adv. Healthcare
[36] R. Riahi, A. Tamayol, S. A. M. Shaegh, A. M. Ghaemmaghami, Mater. 2015, 4, 1915.
M. R. Dokmeci, A. Khademshosseini, Curr. Opin. Chem. Eng. 2015, [70] A. Purnama, H. Hermawan, J. Couet, D. Mantovani, Acta Bio-
7, 101. mater. 2010, 6, 1800.
[37] W. Martanto, J. S. Moore, O. Kashlan, R. Kamath, P. M. Wang, [71] Y. F. Zheng, X. N. Gu, F. Witte, Mater. Sci. Eng., R 2014, 77, 1.
J. M. O’Neal, M. R. Prausnitz, Pharm. Res. 2006, 23, 104. [72] Q. Liu, L. Jiang, R. Shi, L. Zhang, Prog. Polym. Sci. 2012, 37, 715.
[38] G. Novajra, J. Lousteau, D. Milanese, C. Vitale-Brovarone, Mater. [73] L. S. Nair, C. T. Laurencin, Prog. Polym. Sci. 2007, 32, 762.
Lett. 2013, 99, 125. [74] B. D. Ulery, L. S. Nair, C. T. Laurencin, J. Polym. Sci., Part B: Polym.
[39] T. Czuryszkiewicz, J. Ahvenlammi, P. Kortesuo, M. Ahola, F. Kleitz, Phys. 2011, 49, 832.
M. Jokinen, M. Lindén, J. B. Rosenholm, J. Non-Cryst. Solids 2002, [75] J. Intra, J. M. Glasgow, H. Q. Mai, A. K. Salem, J. Controlled Release
306, 1. 2008, 127, 280.
[40] Y. Li, B. Li, G. Xu, Z. Ahmad, Z. Ren, Y. Dong, X. Li, W. Weng, [76] M. Kaur, K. M. Gupta, A. E. Poursaid, P. Karra, A. Mahalingam,
G. Han, Colloids Surf., B 2014, 122, 785. H. A. Aliyar, P. F. Kiser, Drug Delivery Transl. Res. 2011, 1, 223.
[41] J. Xie, E. R. Blough, C. H. Wang, Acta Biomater. 2012, 8, 811. [77] L. K. Petersen, B. Narasimhan, Expert Opin. Drug Delivery 2008, 5,
[42] M. Windbergs, D. A. Weitz, Small 2011, 7, 3011. 837.
[43] P. S. Dittrich, A. Manz, Nat. Rev. Drug Discovery 2006, 5, 210. [78] D. Roy, J. N. Cambre, B. S. Sumerlin, Prog. Polym. Sci. 2010, 35, 278.
[44] H. Gensler, R. Sheybani, P. Y. Li, R. Lo Mann, E. Meng, Biomed. [79] N. Oliva, J. Conde, K. Wang, N. Artzi, Acc. Chem. Res. 2017, 50,
Microdevices 2012, 14, 483. 669.
[45] S. Giri, B. G. Trewyn, M. P. Stellmaker, V. S. Y. Lin, Angew. Chem., [80] M. A. C. Stuart, W. T. S. Huck, J. Genzer, M. Müller, C. Ober,
Int. Ed. 2005, 44, 5038. M. Stamm, G. B. Sukhorukov, I. Szleifer, V. V. Tsukruk, M. Urban,
[46] D. P. Ferris, Y. L. Zhao, N. M. Khashab, H. A. Khatib, J. F. Stoddart, F. Winnik, S. Zauscher, I. Luzinov, S. Minko, Nat. Mater. 2010, 9,
J. I. Zink, J. Am. Chem. Soc. 2009, 131, 1686. 101.
[47] B. Ergün, L. De Cola, H. J. Galla, N. S. Kehr, Adv. Healthcare Mater. [81] S. Mura, J. Nicolas, P. Couvreur, Nat. Mater. 2013, 12, 991.
2016, 5, 1588. [82] C. Park, K. Oh, S. C. Lee, C. Kim, Angew. Chem., Int. Ed. 2007, 46,
[48] Z. Li, J. C. Barnes, A. Bosoy, J. F. Stoddart, J. I. Zink, Chem. Soc. 1455.
Rev. 2012, 41, 2590. [83] X. Jiao, Y. Li, F. Li, W. Wang, Y. Wen, Y. Song, X. Zhang, RSC Adv.
[49] P. Yang, S. Gai, J. Lin, Chem. Soc. Rev. 2012, 41, 3679. 2016, 6, 52292.
[50] H. Qu, S. Bhattacharyya, P. Ducheyne, Adv. Drug Delivery Rev. [84] Y. Yi, A. Zaher, O. Yassine, J. Kosel, I. G. Foulds, Biomicrofluidics
2015, 94, 96. 2015, 9, 52608.
[51] E. Aznar, M. Oroval, L. Pascual, J. R. Murguía, R. Martínez-Mánez, [85] Z. Liu, W. Wang, R. Xie, X.-J. Ju, L.-Y. Chu, Chem. Soc. Rev. 2016,
F. Sancenón, Chem. Rev. 2016, 116, 561. 45, 460.
[52] I. Mylonaki, É. Allémann, F. Saucy, J. A. Haefliger, F. Delie, [86] T. G. Leong, C. L. Randall, B. R. Benson, A. M. Zarafshar,
O. Jordan, Biomaterials 2017, 128, 56. D. H. Gracias, Lab Chip 2008, 8, 1621.
[53] J. A. Lyndon, B. J. Boyd, N. Birbilis, J. Controlled Release 2014, 179, [87] K. Malachowski, J. Breger, H. R. Kwag, M. O. Wang, J. P. Fisher,
63. F. M. Selaru, D. H. Gracias, Angew. Chem., Int. Ed. 2014, 53, 8045.
[54] J. Li, B. Liu, Y. Zhou, Z. Chen, L. Jiang, W. Yuan, L. Liang, PLoS One [88] N. Rapoport, Prog. Polym. Sci. 2007, 32, 962.
2017, 12, 1. [89] C. L. Stevenson, J. T. Santini, R. Langer, Adv. Drug Delivery Rev.
[55] W. J. E. M. Habraken, J. G. C. Wolke, J. A. Jansen, Adv. Drug 2012, 64, 1590.
Delivery Rev. 2007, 59, 234. [90] F. J. Martin, C. Grove, Biomed. Microdevices 2001, 3, 97.
[56] P. Díaz-Rodríguez, M. Landin, A. Rey-Rico, J. Couceiro, T. Coenye, [91] Y. Li, R. S. Shawgo, B. Tyler, P. T. Henderson, J. S. Vogel,
P. González, J. Serra, M. López-Álvarez, B. León, J. Mater. Sci.: A. Rosenberg, P. B. Storm, R. Langer, H. Brem, M. J. Cima, J. Con-
Mater. Med. 2011, 22, 339. trolled Release 2004, 100, 211.
[57] B. Stoeber, D. Liepmann, J. Microelectromech. S. 2005, 14, 472. [92] R. Langer, Science 1990, 249, 1527.
[58] M. C. Peterman, J. Noolandi, M. S. Blumenkranz, H. A. Fishman, [93] C. Wischke, A. T. Neffe, B. D. Hanh, C. F. Kreiner, K. Sternberg,
Proc. Natl. Acad. Sci. USA 2004, 101, 9951. O. Stachs, R. F. Guthoff, A. Lendlein, J. Controlled Release 2013,
[59] L. Lin, A. P. Pisano, J. Microelectromech. S. 1999, 8, 78. 172, 1002.
[60] D. Naumenko, V. Snitka, M. Duch, N. Torras, J. Esteve, Microelec- [94] J.-H. Lee, R. M. Pidaparti, G. M. Atkinson, R. S. Moorthy, J. Drug
tron. Eng. 2012, 98, 488. Delivery 2012, 2012, 1.
[61] H. Zhang, J. K. Jackson, C. Bian, H. M. Burt, M. Chiao, Adv. Mater. [95] B. C. Masi, B. M. Tyler, H. Bow, R. T. Wicks, Y. Xue, H. Brem,
Interfaces 2015, 2, 1. R. Langer, M. J. Cima, Biomaterials 2012, 33, 5768.
[62] H. Zhang, C. Bian, J. K. Jackson, F. Khademolhosseini, H. M. Burt, [96] T. Hoare, J. Santamaria, G. F. Goya, S. Irusta, D. Lin, S. Lau,
M. Chiao, ACS Appl. Mater. Interfaces 2014, 6, 9126. R. Padera, R. Langer, D. S. Kohane, Nano Lett. 2009, 9, 3651.
[63] E. Lee, H. Zhang, J. K. Jackson, C. J. Lim, M. Chiao, RSC Adv. 2016, [97] K. Cai, Z. Luo, Y. Hu, X. Chen, Y. Liao, L. Yang, L. Deng, Adv. Mater.
6, 79900. 2009, 21, 4045.
[64] H. Zhang, M. Chiao, J. Med. Biol. Eng. 2015, 35, 143. [98] A. Nisar, N. Afzulpurkar, B. Mahaisavariya, A. Tuantranont, Sens.
[65] D. Paolino, P. Sinha, M. Fresta, M. Ferrari, Drug Delivery System, Actuators, B 2008, 130, 917.
in Encyclopedia of Medical Devices and Instrumentation, John Wiley [99] M. W. Ashraf, S. Tayyaba, N. Afzulpurkar, Int. J. Mol. Sci. 2011, 12,
& Sons, Inc, Hoboken 2006, p. 437. 3648.
[66] S. S. Lee, P. Hughes, A. D. Ross, M. R. Robinson, Pharm. Res. [100] A. Cobo, R. Sheybani, E. Meng, Adv. Healthcare Mater. 2015, 4,
2010, 27, 2043. 969.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (28 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

[101] N. C. Tsai, C. Y. Sue, Sens. Actuators, A 2007, 134, 555. [140] J. B. Ciolino, T. R. Hoare, N. G. Iwata, I. Behlau, C. H. Dohlman,
[102] S. R. Paul, S. K. Nayak, A. Anis, K. Pal, Polym.-Plast. Technol. Eng. R. Langer, D. S. Kohane, Invest. Ophthalmol. Visual Sci. 2009, 50,
2016, 55, 965. 3346.
[103] S. H. Lee, Y. Bin Lee, B. H. Kim, C. Lee, Y. M. Cho, S.-N. Kim, [141] J. B. Ciolino, S. P. Hudson, A. N. Mobbs, T. R. Hoare, N. G. Iwata,
C. G. Park, Y.-C. Cho, Y. Bin Choy, Nat. Commun. 2017, 8, 15032. G. R. Fink, D. S. Kohane, Invest. Ophthalmol. Visual Sci. 2011, 52,
[104] F. N. Pirmoradi, J. K. Jackson, H. M. Burt, M. Chiao, Lab Chip 6286.
2011, 11, 3072. [142] M. E. Myles, D. M. Neumann, J. M. Hill, Adv. Drug Delivery Rev.
[105] F. N. Pirmoradi, J. K. Jackson, H. M. Burt, M. Chiao, Lab Chip 2005, 57, 2063.
2011, 11, 2744. [143] J. Frucht-Pery, H. Mechoulam, C. S. Siganos, P. Ever-Hadani,
[106] V. Yadav, W. Duan, P. J. Butler, A. Sen, Annu. Rev. Biophys. 2015, 44, M. Shapiro, A. Domb, Exp. Eye Res. 2004, 78, 745.
77. [144] E. Eljarrat-Binstock, A. J. Domb, J. Controlled Release 2006, 110, 479.
[107] E. Meng, T. Hoang, Adv. Drug Delivery Rev. 2012, 64, 1628. [145] Y. Zhang, Y. Chen, M. Yang, X. Yu, Y. Qi, Z. Li, 18th Int.Conf. Solid-
[108] F. E. Kane, J. Burdan, A. Cutino, K. E. Green, Expert Opin. Drug State Sensors, Actuators and Microsystems (Transducers 2015),
Delivery 2008, 5, 1039. Anchorage, Alaska, USA 2015, p. 1041.
[109] P. Zachkani, J. K. Jackson, F. N. Pirmoradi, M. Chiao, RSC Adv. [146] Y. Zhang, Y. Chen, Y. Qi, D. Huang, M. Yang, X. Yu, Y. Hu, Z. Li,
2015, 5, 98087. Sens. Actuators, B 2016, 237, 1007.
[110] A. K. Banga, Expert Opin. Drug Delivery 2009, 6, 343. [147] C. Gupta, A. Chauhan, J. Controlled Release 2011, 150, 70.
[111] V. K. Yellepeddi, R. Sheshala, H. McMillan, C. Gujral, D. Jones, [148] M. N. Yasin, D. Svirskis, A. Seyfoddin, I. D. Rupenthal, J. Controlled
T. Raghu Raj Singh, Drug Discovery Today 2015, 20, 884. Release 2014, 196, 208.
[112] S. Z. Razzacki, P. K. Thwar, M. Yang, V. M. Ugaz, M. A. Burns, Adv. [149] J. Marsh, R. M. Pidaparti, J. Med. Devices 2014, 8, 21005.
Drug Delivery Rev. 2004, 56, 185. [150] N. Haghjou, M. Soheilian, M. J. Abdekhodaie, J. Ophthalmic Vision
[113] B. Koch, I. Rubino, F. S. Quan, B. Yoo, H. J. Choi, Materials 2016, Res. 2011, 6, 317.
9, 646. [151] H. F. Edelhauser, C. L. Rowe-Rendleman, M. R. Robinson,
[114] H. D. Chirra, T. A. Desai, Adv. Drug Delivery Rev. 2012, 64, 1569. D. G. Dawson, G. J. Chader, H. E. Grossniklaus, K. D. Rittenhouse,
[115] B. Ziaie, A. Baldi, M. Lei, Y. Gu, R. A. Siegel, Adv. Drug Delivery Rev. C. G. Wilson, D. A. Weber, B. D. Kuppermann, K. G. Csaky,
2004, 56, 145. T. W. Olsen, U. B. Kompella, V. M. Holers, G. S. Hageman,
[116] M. A. Unger, H. P. Chou, T. Thorsen, A. Scherer, S. R. Quake, Sci- B. C. Gilger, P. A. Campochiaro, S. M. Whitcup, W. T. Wong, Invest.
ence 2000, 288, 113. Ophthalmol. Visual Sci. 2010, 51, 5403.
[117] N. T. Nguyen, S. A. M. Shaegh, N. Kashaninejad, D. T. Phan, Adv. [152] J. Wang, A. Jiang, M. Joshi, J. Christoforidis, Mediators Inflamma-
Drug Delivery Rev. 2013, 65, 1403. tion 2013, 2013, 1.
[118] M. A. Alhnan, T. C. Okwuosa, M. Sadia, K. W. Wan, W. Ahmed, [153] S. A. Molokhia, H. Sant, J. Simonis, C. J. Bishop, R. M. Burr,
B. Arafat, Pharm. Res. 2016, 33, 1817. B. K. Gale, B. K. Ambati, Vision Res. 2010, 50, 680.
[119] N. Sandler, M. Preis, Trends Pharmacol. Sci. 2016, 37, 1070. [154] J. H. Kim, H. B. Song, K. J. Lee, I. H. Seo, J. Y. Lee, S. M. Lee,
[120] G. F. Acosta-Vélez, B. M. Wu, Polym. Sci. 2016, 2, 1. J. H. Kim, W. Ryu, J. Controlled Release 2015, 209, 272.
[121] G. Jonathan, A. Karim, Int. J. Pharm. 2016, 499, 376. [155] J. Jiang, H. S. Gill, D. Ghate, B. E. McCarey, S. R. Patel,
[122] H. L. Quinn, C. M. Hughes, R. F. Donnelly, Int. J. Pharm. 2016, H. F. Edelhauser, M. R. Prausnitz, Invest. Ophthalmol. Visual Sci.
512, 366. 2007, 48, 4038.
[123] S. Mahler, I. Roy, J. Chem. Technol. Biotechnol. 2015, 90, 1167. [156] R. R. Thakur Singh, I. Tekko, K. McAvoy, H. McMillan, D. Jones,
[124] M. J. Cima, H. Lee, K. Daniel, L. M. Tanenbaum, A. Mantzavinou, R. F. Donnelly, Expert Opin. Drug Delivery 2017, 14, 525.
K. C. Spencer, Q. Ong, J. C. Sy, J. Santini, C. M. Schoellhammer, [157] O. Khandan, A. Famili, M. Y. Kahook, M. P. Rao, Engineering in
D. Blankschtein, R. S. Langer, J. Controlled Release 2014, 190, 157. Medicine and Biology Society (EMBC), in 34th Annual Int. Conf. of
[125] N. Gooch, S. A. Molokhia, R. Condie, R. M. Burr, B. Archer, the IEEE EMBS, San Diego, USA 2012, p. 6572.
B. K. Ambati, B. Wirostko, Pharmaceutics 2012, 4, 197. [158] O. Khandan, M. Y. Kahook, M. P. Rao, Sens. Actuators, B 2016,
[126] S. S. Lee, M. R. Robinson, Ophthalmic Res. 2009, 41, 124. 223, 15.
[127] J. Nirmal, K. Radhakrishnan, M. Moreno, J. V. Natarajan, A. Laude, [159] S. R. Patel, D. E. Berezovsky, B. E. McCarey, V. Zarnitsyn,
T. H. Lim, S. Venkatraman, R. Agrawal, Expert Opin. Drug Delivery H. F. Edelhauser, M. R. Prausnitz, Invest. Ophthalmol. Visual Sci.
2016, 13, 1625. 2012, 53, 4433.
[128] E. Lavik, M. H. Kuehn, Y. H. Kwon, Eye 2011, 25, 578. [160] S. R. Patel, A. S. P. Lin, H. F. Edelhauser, M. R. Prausnitz, Pharm.
[129] D. Achouri, K. Alhanout, P. Piccerelle, V. Andrieu, Drug Dev. Ind. Res. 2011, 28, 166.
Pharm. 2013, 39, 1599. [161] J. Jiang, J. S. Moore, H. F. Edelhauser, M. R. Prausnitz, Pharm. Res.
[130] A. Urtti, Adv. Drug Delivery Rev. 2006, 58, 1131. 2009, 26, 395.
[131] D. Ghate, H. F. Edelhauser, Expert Opin. Drug Delivery 2006, 3, [162] B. Chiang, Y. C. Kim, H. F. Edelhauser, M. R. Prausnitz, Exp. Eye
275. Res. 2016, 145, 424.
[132] D. H. Geroski, H. F. Edelhauser, Invest. Ophthalmol. Visual Sci. [163] B. C. Gilger, E. M. Abarca, J. H. Salmon, S. Patel, Invest. Oph-
2000, 41, 961. thalmol. Visual Sci. 2013, 54, 2483.
[133] H. Chen, J. Drug Targeting 2015, 23, 597. [164] N. Nagai, H. Kaji, H. Onami, Y. Ishikawa, M. Nishizawa,
[134] R. Gaudana, H. K. Ananthula, A. Parenky, A. K. Mitra, AAPS J. N. Osumi, T. Nakazawa, T. Abe, Acta Biomater. 2014, 10, 680.
2010, 12, 348. [165] R. Lo, K. Kuwahara, P. Y. Li, R. Agrawal, M. S. Humayun, E. Meng,
[135] B. N. Kuno, S. Fujii, Retina Today 2012, May/June, 54. Int. Conf. Microtechnol. Med. Biol. Okinawa, Japan 2006, p. 74.
[136] N. Kuno, S. Fujii, Polymers 2011, 3, 193. [166] R. Lo, P.-Y. Li, S. Saati, R. Agrawal, M. S. Humayun, E. Meng, Lab
[137] D. Lee, S. Cho, H. S. Park, I. Kwon, Sci. Rep. 2016, 6, 34194. Chip 2008, 8, 1027.
[138] L. Xinming, C. Yingde, A. W. Lloyd, S. V. Mikhalovsky, [167] R. Lo, P. Y. Li, S. Saati, R. N. Agrawal, M. S. Humayun, E. Meng,
S. R. Sandeman, C. A. Howel, L. Liewen, Contact Lens Anterior Eye Biomed. Microdevices 2009, 11, 959.
2008, 31, 57. [168] P. Y. Li, J. Shih, R. Lo, S. Saati, R. Agrawal, M. S. Humayun,
[139] C.-M. Phan, Clin. Exp. Pharmacol. 2013, 4, 1. Y. C. Tai, E. Meng, Sens. Actuators, A 2008, 143, 41.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (29 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

[169] S. Saati, R. Lo, P.-Y. Li, E. Meng, R. Varma, M. S. Humayun, Curr. [202] M. Quirini, A. Menciassi, S. Scapellato, P. Dario, F. Rieber,
Eye Res. 2010, 35, 192. C. N. Ho, S. Schostek, M. O. Schurr, Gastrointest. Endosc. 2008, 67,
[170] P. Song, S. Kuang, N. Panwar, G. Yang, D. J. H. Tng, S. C. Tjin, 1153.
W. J. Ng, M. B. A. Majid, G. Zhu, K. T. Yong, Z. L. Wang, Adv. [203] M. Quirini, A. Menciassi, S. Scapellato, C. Stefanini, P. Dario,
Mater. 2017, 29, 1. IEEE/ASME Trans. Mechatronics 2008, 13, 169.
[171] M. Humayun, A. Santos, J. C. Altamirano, R. Ribeiro, R. Gonzalez, [204] S. P. Woods, T. G. Constandinou, S. Member, IEEE Trans. Biomed.
A. de la Rosa, J. Shih, C. Pang, F. Jiang, P. Calvillo, J. Huculak, Eng. 2013, 60, 945.
J. Zimmerman, S. Caffey, Transl. Vision Sci. Technol. 2014, 3, 5. [205] P. Valdastri, R. J. Webster, C. Quaglia, M. Quirini, A. Menciassi,
[172] J.-C. Gutierrez-Hernandez, S. Caffey, W. Abdallah, P. Dario, IEEE Trans. Rob. 2009, 25, 1047.
P. Calvillo, R. Gonzalez, J. Shih, J. Brennan, J. Zimmerman, [206] S. P. Woods, T. G. Constandinou, IEEE Eng. Med. Biol. Soc. Conf.
J.-C. Martinez-Camarillo, A. R. Rodriguez, R. Varma, A. Santos, 2011, 2011, 7372.
G. Sanchez, M. Humayun, Transl. Vision Sci. Technol. 2014, 3, 1. [207] H. Richert, O. Surzhenko, S. Wangemann, J. Heinrich, P. Görnert,
[173] F. N. Pirmoradi, K. Ou, J. K. Jackson, K. Letchford, J. Cui, J. Magn. Magn. Mater. 2005, 293, 497.
K. T. Wolf, F. Graber, T. Zhao, J. A. Matsubara, H. Burt, M. Chiao, [208] C. T. Dietzel, H. Richert, S. Abert, U. Merkel, M. Hippius,
L. Lin, 26th Int. Conf. Micro Electro Mech. Syst. (MEMS), Taipei, A. Stallmach, J. Controlled Release 2012, 161, 722.
Taiwan 2013, p. 1. [209] S. Yim, M. Sitti, IEEE Trans. Rob. 2012, 28, 183.
[174] S. Sant, S. L. Tao, O. Z. Fisher, Q. Xu, N. A. Peppas, [210] S. Yim, K. Goyal, M. Sitti, IEEE/ASME Trans. Mechatronics 2013,
A. Khademhosseini, Adv. Drug Delivery Rev. 2012, 64, 496. 18, 1413.
[175] C. B. Fox, J. Kim, L. V. Le, C. L. Nemeth, H. D. Chirra, T. A. Desai, J. [211] C. L. Randall, T. G. Leong, N. Bassik, D. H. Gracias, Adv. Drug
Controlled Release 2015, 219, 431. Delivery Rev. 2007, 59, 1547.
[176] Z. Shen, S. Mitragotri, Pharm. Res. 2002, 19, 391. [212] R. Fernandes, D. H. Gracias, Adv. Drug Delivery Rev. 2012, 64, 1579.
[177] K. Whitehead, Z. Shen, S. Mitragotri, J. Controlled Release 2004, 98, [213] T. Leong, Z. Gu, T. Koh, D. H. Gracias, J. Am. Chem. Soc. 2006,
37. 128, 11336.
[178] V. Gupta, B. H. Hwang, J. Lee, A. C. Anselmo, N. Doshi, [214] H. Ye, C. L. Randall, T. G. Leong, D. A. Slanac, E. K. Call,
S. Mitragotri, J. Controlled Release 2013, 172, 753. D. H. Gracias, Angew. Chem., Int. Ed. 2007, 46, 4991.
[179] A. Ahmed, C. Bonner, T. A. Desai, Biomed. Microdevices 2001, 3, 89. [215] M. R. Prausnitz, R. Langer, Nat. Biotechnol. 2008, 26, 1261.
[180] A. Ahmed, C. Bonner, T. A. Desai, J. Controlled Release 2002, 81, [216] M. R. Prausnitz, S. Mitragotri, R. Langer, Nat. Rev. Drug Discovery
291. 2004, 3, 115.
[181] S. L. Tao, T. A. Desai, Adv. Mater. 2005, 17, 1625. [217] M. Ochoa, C. Mousoulis, B. Ziaie, Adv. Drug Delivery Rev. 2012, 64, 1603.
[182] S. L. Tao, T. A. Desai, J. Controlled Release 2005, 109, 127. [218] A. Arora, M. R. Prausnitz, S. Mitragotri, Int. J. Pharm. 2008, 364, 227.
[183] K. M. Ainslie, C. M. Kraning, T. A. Desai, Lab Chip 2008, 8, 1042. [219] M. R. Prausnitz, Adv. Drug Delivery Rev. 2004, 56, 581.
[184] K. M. Ainslie, R. D. Lowe, T. T. Beaudette, L. Petty, E. M. Bachelder, [220] R. K. Sivamani, D. Liepmann, H. I. Maibach, Expert Opin. Drug
T. A. Desai, Small 2009, 5, 2857. Delivery 2007, 4, 19.
[185] H. D. Chirra, T. A. Desai, Small 2012, 8, 3839. [221] S. H. Bariya, M. C. Gohel, T. A. Mehta, O. P. Sharma, J. Pharm.
[186] H. D. Chirra, L. Shao, N. Ciaccio, C. B. Fox, J. M. Wade, A. Ma, Pharmacol. 2012, 64, 11.
T. A. Desai, Adv. Healthcare Mater. 2014, 3, 1648. [222] R. F. Donnelly, T. R. R. Singh, A. D. Woolfson, Drug Delivery 2010,
[187] C. B. Fox, Y. Cao, C. L. Nemeth, H. D. Chirra, R. W. Chevalier, 17, 187.
A. M. Xu, N. A. Melosh, T. A. Desai, ACS Nano 2016, 10, 5873. [223] Y. C. Kim, J. H. Park, M. R. Prausnitz, Adv. Drug Delivery Rev. 2012,
[188] M. Rasouli, L. Lin, A. P. Kencana, K.-J. Wong, S.-L. Tan, K. Y. Ho, 64, 1547.
S. J. Phee, J. Healthcare Eng. 2011, 2, 459. [224] K. van der Maaden, W. Jiskoot, J. Bouwstra, J. Controlled Release
[189] F. Munoz, G. Alici, W. Li, Adv. Drug Delivery Rev. 2014, 71, 77. 2012, 161, 645.
[190] C. Quaglia, S. Tognarelli, E. Sinibaldi, N. Funaro, P. Dario, [225] H. S. Gill, D. D. Denson, B. A. Burris, M. R. Prausnitz, Clin. J. Pain
A. Menciassi, J. Med. Devices 2013, 8, 14503. 2008, 24, 585.
[191] P. J. van der Schaar, J. F. Dijksman, H. Broekhuizen-De Gast, [226] W. Martanto, S. Davis, N. R. Holiday, J. Wang, H. Gill,
J. Shimizu, N. Van Lelyveld, H. Zou, V. Iordanov, C. Wanke, M. Prausnitz, Pharm. Res. 2004, 21, 947.
P. D. Siersema, Gastrointest. Endosc. 2013, 78, 520. [227] H. S. Gill, M. R. Prausnitz, Pharm. Res. 2007, 24, 1369.
[192] E. Söderlind, B. Abrahamsson, F. Erlandsson, C. Wanke, [228] Y. C. Kim, F. S. Quan, R. W. Compans, S. M. Kang, M. R. Prausnitz,
V. Iordanov, C. von Corswant, J. Controlled Release 2015, 217, 300. J. Controlled Release 2010, 142, 187.
[193] M. E. A. McGirr, S. M. McAllister, E. E. Peters, A. W. Vickers, [229] M. Han, D. K. Kim, S. H. Kang, H. R. Yoon, B. Y. Kim, S. S. Lee,
A. F. Parr, A. W. Basit, Eur. J. Pharm. Sci. 2009, 36, 386. K. D. Kim, H. G. Lee, Sens. Actuators, B 2009, 137, 274.
[194] H. N. E. Stevens, C. G. Wilson, P. G. Welling, M. Bakhshaee, [230] K. Fukushima, A. Ise, H. Morita, R. Hasegawa, Y. Ito, N. Sugioka,
J. S. Binns, A. C. Perkins, M. Frier, E. P. Blackshaw, M. W. Frame, K. Takada, Pharm. Res. 2011, 28, 7.
D. J. Nichols, M. J. Humphrey, S. R. Wicks, Int. J. Pharm. 2002, [231] S. P. Sullivan, D. G. Koutsonanos, M. del Pilar Martin, J. W. Lee,
236, 27. V. Zarnitsyn, S.-O. Choi, N. Murthy, R. W. Compans, I. Skountzou,
[195] I. R. Wilding, A. L. Connor, P. Carpenter, C. Rordorf, J. Branson, M. R. Prausnitz, Nat. Med. 2010, 16, 915.
S. Milosavljev, G. Scott, Pharm. Res. 2004, 21, 443. [232] S. P. Davis, W. Martanto, M. G. Allen, M. R. Prausnitz, IEEE Trans.
[196] S. Murad, J. Murad, H. Khan, Rawal Med. J. 2013, 38, 66. Biomed. Eng. 2005, 52, 909.
[197] J. Cui, X. Zheng, W. Hou, Y. Zhuang, X. Pi, J. Yang, Telemed. J. [233] F. Pérennès, B. Marmiroli, M. Matteucci, M. Tormen, L. Vaccari,
e-health 2008, 14, 715. E. Di Fabrizio, J. Micromech. Microeng. 2006, 16, 473.
[198] R. Gröning, H. Bensmann, R. S. Müller, Int. J. Pharm. 2008, 364, 9. [234] S. A. Burton, C. Y. Ng, R. Simmers, C. Moeckly, D. Brandwein,
[199] R. Gröning, H. Bensmann, Eur. J. Pharm. Biopharm. 2009, 72, 282. T. Gilbert, N. Johnson, K. Brown, T. Alston, G. Prochnow,
[200] P. Xitian, L. Hongying, W. Kang, L. Yulin, Z. Xiaolin, W. Zhiyu, Int. K. Siebenaler, K. Hansen, Pharm. Res. 2011, 28, 31.
J. Pharm. 2009, 382, 160. [235] S. Y. Yang, E. D. O’Cearbhaill, G. C. Sisk, K. M. Park, W. K. Cho,
[201] X. Pi, Y. Lin, K. Wei, H. Liu, G. Wang, X. Zheng, Z. Wen, D. Li, M. Villiger, B. E. Bouma, B. Pomahac, J. M. Karp, Nat. Commun.
Sens. Actuators, A 2010, 159, 227. 2013, 4, 1702.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (30 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

[236] H. Wang, G. Pastorin, C. Lee, Adv. Sci. 2016, 3, 1. [262] A. Altuna, E. Bellistri, E. Cid, P. Aivar, B. Gal, J. Berganzo,
[237] L. Y. Chu, M. R. Prausnitz, J. Controlled Release 2011, G. Gabriel, A. Guimerà, R. Villa, L. J. Fernández,
149, 242. L. Menendez de la Prida, Lab Chip 2013, 13, 1422.
[238] D. D. Zhu, Q. L. Wang, X. B. Liu, X. D. Guo, Acta Biomater. 2016, [263] R. R. Zhang, J. A. Lubin, J. S. Kuo, Neurosurgery 2016, 79, N19.
41, 312. [264] S. Spieth, A. Schumacher, T. Holtzman, P. D. Rich, D. E. Theobald,
[239] U. O. Häfeli, A. Mokhtari, D. Liepmann, B. Stoeber, Biomed. Micro- J. W. Dalley, R. Nouna, S. Messner, R. Zengerle, Biomed. Microde-
devices 2009, 11, 943. vices 2012, 14, 799.
[240] H. Lee, T. K. Choi, Y. B. Lee, H. R. Cho, R. Ghaffari, L. Wang, [265] N. El Kechai, F. Agnely, E. Mamelle, Y. Nguyen, E. Ferrary,
H. J. Choi, T. D. Chung, N. Lu, T. Hyeon, S. H. Choi, D.-H. Kim, A. Bochot, Int. J. Pharm. 2015, 494, 83.
Nat. Nanotechnol. 2016, 11, 566. [266] H. Staecker, C. Jolly, C. Garnham, Drug Discovery Today 2010, 15, 314.
[241] M. Kim, J. An, K. S. Kim, M. Choi, M. Humar, S. J. J. Kwok, T. Dai, [267] J. T. Borenstein, Expert Opin. Drug Delivery 2011, 8, 1161.
S. H. Yun, Biomed. Opt. Express 2016, 7, 4220. [268] W. F. Sewell, J. T. Borenstein, Z. Chen, J. Fiering, O. Handzel,
[242] B. Kim, S. Yoo, Y. J. Kim, J. Park, B. Kang, S. Haam, S. W. Kang, M. Holmboe, E. S. Kim, S. G. Kujawa, M. J. McKenna,
K. Kang, U. Jeong, Adv. Mater. Interfaces 2016, 3, 1. M. M. Mescher, B. Murphy, E. E. Eary Swan, M. Peppi, S. Tao,
[243] Y. Liu, A. Servant, O. J. Guy, K. T. Al-Jamal, P. R. Williams, Audiol. Neurotol. 2009, 14, 411.
K. M. Hawkins, K. Kostarelos, Sens. Actuators, B 2012, 175, 100. [269] J. Fiering, M. J. Mescher, E. E. Leary Swan, M. E. Holmboe,
[244] W. H. Ryu, M. Vyakarnam, R. S. Greco, F. B. Prinz, R. J. Fasching, B. A. Murphy, Z. Chen, M. Peppi, W. F. Sewell, M. J. McKenna,
Biomed. Microdevices 2007, 9, 845. S. G. Kujawa, J. T. Borenstein, Biomed. Microdevices 2009, 11, 571.
[245] S. Ferrati, D. Fine, J. You, E. De Rosa, L. Hudson, E. Zabre, [270] E. S. Kim, E. Gustenhoven, M. J. Mescher, E. E. Leary Pararas,
S. Hosali, L. Zhang, C. Hickman, S. Sunder Bansal, K. A. Smith, A. J. Spencer, V. Tandon, J. T. Borenstein, J. Fiering,
A. M. Cordero-Reyes, T. Geninatti, J. Sih, R. Goodall, G. Palapattu, Lab Chip 2014, 14, 710.
M. Kloc, R. M. Ghobrial, M. Ferrari, A. Grattoni, J. Controlled [271] O. A. Scholz, A. Wolff, A. Schumacher, L. I. Giannola, G. Campisi,
Release 2013, 172, 1011. T. Ciach, T. Velten, Drug Discovery Today 2008, 13, 247.
[246] S. C. Sundararaj, M. V. Thomas, R. Peyyala, T. D. Dziubla, [272] A. Schumacher, T. Goettsche, S. Haeberle, T. Velten, O. Scholz,
D. A. Puleo, Biomaterials 2013, 34, 8835. A. Wolff, B. Beiski, S. Messner, R. Zengerle, IFMBE Proc. 2008, 22,
[247] S. C. Sundararaj, M. Al-Sabbagh, C. L. Rabek, T. D. Dziubla, 2352.
M. V. Thomas, D. A. Puleo, J. Biomed. Mater. Res., Part B 2016, [273] C. Paderni, G. Campisi, A. Schumacher, T. Göttsche, L. I. Giannola,
104, 1302. V. De Caro, A. Wolff, Int. J. Pharm. 2013, 452, 128.
[248] Y. Li, H. L. H. Duc, B. Tyler, T. Williams, M. Tupper, R. Langer, [274] K. J. Whaley, J. Hanes, R. Shattock, R. A. Cone, D. R. Friend, Anti-
H. Brem, M. J. Cima, J. Controlled Release 2005, 106, 138. viral Res. 2010, 88, S55.
[249] J. M. Maloney, S. A. Uhland, B. F. Polito, N. F. Sheppard, [275] A. Bernkop-schn, M. Hornof, Am. J. Drug Delivery 2003, 1, 241.
C. M. Pelta, J. T. Santini, J. Controlled Release 2005, 109, 244. [276] A. Hussain, F. Ahsan, J. Controlled Release 2005, 103, 301.
[250] J. H. Prescott, S. Lipka, S. Baldwin, N. F. Sheppard, J. M. Maloney, [277] R. Karl Malcolm, S. M. Fetherston, C. F. McCoy, P. Boyd, I. Major,
J. Coppeta, B. Yomtov, M. A. Staples, J. T. Santini, Nat. Biotechnol. Int. J. Women’s Health 2012, 4, 595.
2006, 24, 437. [278] P. Boyd, S. M. Fetherston, C. F. McCoy, I. Major, D. J. Murphy,
[251] J. H. Prescott, T. J. Krieger, S. Lipka, M. A. Staples, Pharm. Res. S. Kumar, J. Holt, A. Brimer, W. Blanda, B. Devlin, R. K. Malcolm,
2007, 24, 1252. Int. J. Pharm. 2016, 511, 619.
[252] E. R. Proos, J. H. Prescott, M. A. Staples, Pharm. Res. 2008, 25, [279] Y. Chen, Y. L. Traore, A. Li, K. R. Fowke, E. A. Ho, Drug Des., Dev.
1387. Ther. 2014, 8, 1801.
[253] R. Farra, N. F. Sheppard, L. McCabe, R. M. Neer, J. M. Anderson, [280] R. J. Morrow, A. D. Woolfson, L. Donnelly, R. Curran, G. Andrews,
J. T. Santini, M. J. Cima, R. Langer, Sci. Transl. Med. 2012, 4, D. Katinger, R. K. Malcolm, Eur. J. Pharm. Biopharm. 2011, 77, 3.
122ra21. [281] R. S. Teller, R. Rastogi, T. J. Johnson, M. J. Blair, R. W. Hitchcock,
[254] T. Hoare, B. P. Timko, J. Santamaria, G. F. Goya, S. Irusta, S. Lau, P. F. Kiser, Pharm. Res. 2014, 31, 2344.
C. F. Stefanescu, D. Lin, R. Langer, D. S. Kohane, Nano Lett. 2011, [282] S. Rose, A. Chaudhari, C. M. Peterson, Adv. Drug Delivery Rev.
11, 1395. 2009, 61, 808.
[255] O. Ordeig, S. Y. Chin, S. Kim, P. V. Chitnis, S. K. Sia, Sci. Rep. 2016, [283] X. Zhou, Y. Li, X. Jiang, L. Qiu, J. Liu, Eur. J. Contracept. Reprod.
6, 22803. Health Care 2010, 15, 205.
[256] J. Shi, H. Zhang, J. Jackson, A. Shademani, M. Chiao, J. Mater. [284] D. Wildemeersch, Contraception 2007, 75, S82.
Chem. B 2016, 4, 7415. [285] X. Xia, Y. Tang, C. Xie, Y. Wang, S. Cai, C. Zhu, J. Mater. Sci.: Mater.
[257] A. Shademani, H. Zhang, J. K. Jackson, M. Chiao, Adv. Funct. Med. 2011, 22, 1773.
Mater. 2017, 27, 1. [286] J. Xi, X. A. Si, R. Gaide, PLoS One 2014, 9, e86593.
[258] S. Metz, A. Bertsch, D. Bertrand, P. Renaud, Biosens. Bioelectron. [287] J. E. Lee, S. Park, M. Park, M. H. Kim, C. G. Park, S. H. Lee,
2004, 19, 1309. S. Y. Choi, B. H. Kim, H. J. Park, J. H. Park, C. Y. Heo, Y. Bin Choy,
[259] S. Takeuchi, D. Ziegler, Y. Yoshida, K. Mabuchi, T. Suzuki, Lab Chip Acta Biomater. 2013, 9, 8318.
2005, 5, 519. [288] V. Jannin, G. Lemagnen, P. Gueroult, D. Larrouture, C. Tuleu, Adv.
[260] K. Seidl, S. Spieth, S. Herwik, J. Steigert, R. Zengerle, O. Paul, Drug Delivery Rev. 2014, 73, 34.
P. Ruther, J. Micromech. Microeng. 2010, 20, 105006. [289] H. Shiohira, M. Fujii, N. Koizumi, M. Kondoh, Y. Watanabe, Int. J.
[261] K. Gao, G. Li, L. Liao, J. Cheng, J. Zhao, Y. Xu, Sens. Actuators, A Pharm. 2009, 379, 119.
2013, 197, 9. [290] M. Staples, WIREs Nanomed. Nanobiotechnol. 2010, 2, 400.

Adv. Funct. Mater. 2017, 27, 1703606 1703606  (31 of 31) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like