COREL-07177; No of Pages 13
Journal of Controlled Release xxx (2014) xxx–xxx
Contents lists available at ScienceDirect
Journal of Controlled Release
journal homepage: www.elsevier.com/locate/jconrel
4
5
6
7
8
9
a
1 0
a r t i c l e
11
21
12
22
13
14
Article history:
Received 26 February 2014
Accepted 2 May 2014
Available online xxxx
15
16
17
18
19
20
Keywords:
Organ-on-a-chip
Drug delivery
Nanoparticle
Drug screening
Toxicity
R
O
O
F
3
Nupura S. Bhise a,b, João Ribas a,b,c,d, Vijayan Manoharan a,b, Yu Shrike Zhang a,b, Alessandro Polini a,b,
Solange Massa a,b, Mehmet R. Dokmeci a,b, Ali Khademhosseini a,b,e,f,⁎
Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA
Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
d
Biocant — Biotechnology Innovation Center, 3060-197 Cantanhede, Portugal
e
Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, USA
f
Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
b
c
i n f o
a b s t r a c t
P
2Q1
Organ-on-a-chip platforms for studying drug delivery systems
Novel microfluidic tools allow new ways to manufacture and test drug delivery systems. Organ-on-a-chip
systems — microscale recapitulations of complex organ functions — promise to improve the drug development
pipeline. This review highlights the importance of integrating microfluidic networks with 3D tissue engineered
models to create organ-on-a-chip platforms, able to meet the demand of creating robust preclinical screening
models. Specific examples are cited to demonstrate the use of these systems for studying the performance of
drug delivery vectors and thereby reduce the discrepancies between their performance at preclinical and clinical
trials. We also highlight the future directions that need to be pursued by the research community for these proofof-concept studies to achieve the goal of accelerating clinical translation of drug delivery nanoparticles.
© 2014 Published by Elsevier B.V.
C
T
E
D
1
33
31
E
32
1. Introduction
35
36
The rapidly developing field of nanomedicine can significantly impact human disease therapy [1,2]. The research progress accomplished
in this field, over the last few decades, has led to the development of
nanomaterials, useful for designing carriers that deliver therapeutic
payload to diseased cells. An ideal drug delivery system should be
easy to manufacture and scale-up, low cost, biocompatible, biodegradable, possesses a high drug loading capacity and can be targeted to the
site-of-interest in the body. Nanocarriers, also routinely referred to as
nanoparticles, are a class of drug delivery systems that range in size
from about 50 to 200 nm, allowing them to efficiently translocate across
the cell membrane barrier.
From a therapeutic standpoint, nanocarriers can prolong the
systemic circulation time of the drug and significantly reduce adverse
side effects caused by off-target delivery at healthy tissue sites. This controlled release of drugs reduces the magnitude of overall drug exposure
required for a therapeutic effect, thus avoiding higher drug doses and
consequent adverse effects. A wide variety of drugs, including hydrophobic and hydrophilic small molecules, as well as biomacromolecules,
43
44
45
46
47
48
49
50
51
52
R
O
C
41
Q2
42
N
39
40
U
37
38
R
34
⁎ Corresponding author at: Division of Biomedical Engineering, Department of
Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139,
USA.
E-mail address: alik@rics.bwh.harvard.edu (A. Khademhosseini).
23
24
25
26
27
28
29
30
can be encapsulated within nanoparticles by tailoring the chemistry of
nanomaterials, polymeric or inorganic/metallic, to achieve the desired
encapsulation capability and release kinetics. The first use of nanoscale
systems for drug delivery was reported in the 1970s, when liposomal
Trojan horse nanoparticles were used for treating lysosomal storage
disease [3,4]. Nanoparticles have also been developed as diagnostic
agents to enhance the sensitivity for imaging techniques, including
X-ray computed tomography (CT) and magnetic resonance imaging
(MRI). An increase in available techniques to engineer more precise
and sophisticated nanomaterials, and a deeper understanding of disease
biology have catapulted a new generation of nanotherapeutics with
improved properties.
The above-mentioned advantages make nanoscale drug delivery
systems appealing to the pharmaceutical companies and healthcare
regulatory agencies. However, in spite of these rapid bench-side developments, the translation of therapeutic nanoparticles to the commercial
pipeline has been less impressive [5]. Very few systems have been
approved by the Food and Drug Administration (FDA), including
Doxil, a liposomal formulation encapsulating the chemotherapeutic
drug Doxorubicin, and Abraxane, based on the nanoparticle albuminbound (nab) technology to deliver Paclitaxel, a widely used drug for
breast and pancreatic cancer [6]. This slow pace of bench-to-bedside
translation can be attributed to several challenges, the most critical
being the lack of robust preclinical tissue culture platforms that can
mimic in vivo conditions and predict the performance of these nanoparticles within the human body.
http://dx.doi.org/10.1016/j.jconrel.2014.05.004
0168-3659/© 2014 Published by Elsevier B.V.
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
77
78
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
C
105
106
E
103
104
R
101
102
R
F
94
95
O
92
93
C
90
91
U
N
88
89
O
Several parameters need to be studied for developing nanoparticles
for clinical use. These include studying the fate of the nanoparticles
inside the body and its toxicological effects, the mode of binding and
internalization at the cellular level, the stability of the nanoparticles
with respect to various physical and chemical conditions of the body,
and, most importantly, the efficacy when compared to free drugs [5].
Large-batch synthesis, toxicity assessment and efficacy screening are
the major levels at which clinical translation of nanotherapeutics faces
set-back [16]. On the manufacturing front, scaling the small lab synthesis techniques to the large-scale production of nanoparticles has been
challenging for the pharmaceutical companies [8]. Meanwhile, screening for the toxicity and efficacy suffers from the paucity of preclinical
models that would robustly predict the nanoparticles' behavior
inside the human body [16]. For simultaneous evaluation of the
above-mentioned parameters, predictive in vitro platforms are essential
while developing drug delivery vectors [13,17].
The current gold standard for preclinical testing of nanotherapeutics
is in vivo studies. These do not accurately predict human responses
due to inter-species difference in genetic makeup, along with being
extremely time-consuming, expensive, low-throughput and raising
ethical concerns. The resolution for whole-animal imaging methods is
limited, hindering visualization during transport of the theranostic
agents in the target tissue. Being unable to reproduce its preclinical performance, many drug delivery systems which pass the preclinical phase
fail to address the toxicity and efficacy effects when compared to their
free drug counterparts in human clinical trials [5]. Strikingly, the main
reason cited for this effect is the use of animal models for optimization
during drug carrier design [5], which brings back the obvious drawback
of a certain degree of physiological irrelevance between human and
animal models.
Animal models need to be complemented with sophisticated in vitro
platforms to fill this gap. In current in vitro studies, drug delivery carriers
are commonly tested in two-dimensional (2D) monolayer cell culture
models. These 2D cultures involve growing on top of a flat substrate
(e.g., glass or polystyrene) a monolayer of single or multiple cell types
that are either freshly isolated from human/animal tissues (primary
cells) or are already established, immortalized cell lines. In these setups,
drug delivery systems are usually mixed with culture media and
directly applied on the cell monolayers, after which cellular responses
are recorded. Among several published studies [18–21], the work of
Xia and colleagues on the cellular uptake of gold nanoparticles
(AuNPs) by SK-BR-3 breast tumor cells [22], stands out by devising a
novel testing method. After culturing the cells on a piece of glass, the
86
87
R
O
100
85
P
2. Limitations of current culture platforms used for developing drug
delivery systems
83
84
substrate was carefully reversed and placed upside down before
AuNPs, with different shapes and sizes, were added in the culture
media. Such an approach successfully avoided the issues caused by
rapid sedimentation of nanoparticles. Indeed, the amount of cellular
uptake of nanoparticles in upright and inverted cultures was found
to significantly depend on the rate of diffusion/sedimentation of the
nanoparticles.
In spite of these novel approaches for 2D cell culture, it is gradually
realized that there are many shortcomings with these “flat” models to
mimic the complex three-dimensional (3D) in vivo microenvironment,
wherein the cells and extracellular matrix (ECM) exist in wellorganized architectures. Moreover, the nanoparticle delivery efficacy
differs considerably between 2D and 3D culture platforms [23]. Primary
cells usually have a limited lifespan, undergo rapid phenotypic alterations, and show large variability over different batches of isolation;
on the other hand, although established cell lines are more stable,
many times they do not present genuine tissue-specific functions [24].
In this regard, efforts were shifted toward developing multiple 3D culture systems that can better recapitulate in vivo tissue functions. Multicellular spheroids are important 3D models for researchers [25–29].
These spheroids are formed by spontaneous aggregation of multiple
cells held together by ECM secreted by residing cells. The apoptotic/
necrotic core of the spheroids contrasts with the proliferative cell layers
on the periphery, providing a better mimic of in vivo tumor environment. Due to the importance and long-time usage of multicellular
spheroids in both pharmaceutical studies and regenerative medicine,
researchers have developed sophisticated methods that allow efficient
fabrication of uniform spheroids at relatively large scales, including
the use of hanging drops, non-adhesive microwells, rotation cultures,
or 3D porous scaffolds [30–37]. Multicellular tumor cylindroids have
been used to study the effect of charge on the uptake of fluorescein isothiocyanate (FITC) or doxorubicin (DOX)-conjugated AuNPs loaded
with drugs, where diffusion is permitted only from the periphery to
the center [38]. Kotov and co-workers directly utilized tumor spheroids
for toxicity testing of CdTe quantum dots and AuNPs [39]. The toxic
effects of these nanoparticles were compared with conventional 2D
cultures, to reveal different responses of cells in terms of morphology,
particle distribution, membrane integrity, mitochondrial activity, and
apoptosis (Fig. 1).
Besides multicellular spheroids, hydrogels and porous scaffolds have
also been widely employed for constructing 3D tissue models at larger
size scales [40–42]. There are a number of advantages associated with
3D cultures within a matrix. For example, the mechanical properties of
the gels can be precisely modulated, which have been shown to determine the phenotypic behaviors of the cells [43–45]; the matrices can
be fabricated to possess various hierarchical structures and any desired
shape to accommodate specific target tissues. As an example, Huang
and co-workers demonstrated that cancer cells became more tumorigenic when cultured in a fibrin gel with a stiffness of approximately
90 Pa, as shown by in vivo tumor formation in mice even when only
very few (10 or 100) tumor cells were injected, whereas the same number of tumor cells from stiff 2D substrates could not induce the formation of tumors [46]. Moreover, Mooney et al. cultured OSCC-3 oral
squamous carcinoma cells within porous poly(lactide-co-glycolide)
(PLG) scaffolds to create an in vitro tumor model [47]. They argued
that tumor cells cultured in PLG scaffolds could better recapitulate
their in vivo states than in 3D Matrigel or 2D substrates as shown by
their morphological appearances, proliferation rates, distribution of
oxygen concentrations, and secretion patterns of biomolecules.
Although static culture systems based on multicellular spheroids or
3D matrices can recapitulate the in vivo functionality of tissues much
better than 2D cultures, they fail to present dynamic flow conditions
that the cells usually experience in the body. The absence of homogenous perfusion results in improper gas and nutrient exchange through
the core of the constructs. Additionally, the gravitational settling of
nanoparticles in static conditions affects the outcome of dosage
D
98
99
81
82
T
96
97
The development of microfluidic platforms for nanoparticle synthesis has shown to overcome several disadvantages of the traditional bulk
synthesis methods such as scalability and batch-to-batch variability
[7–9]. Microfluidic approaches have also been used as a tool for more
sophisticated, faster and highly efficient characterization of the biophysical properties of nanoparticles [10,11]. Additionally, the application of
microfabrication techniques to tissue engineering aided in the creation
of physiologically relevant disease models. Establishment of these techniques has paved the way for robust advances in tissue culture systems
integrated with microfluidic networks [12]. More recently, the demand
for high-throughput drug screening platforms with better preclinical
predictability has translated into major developments in the organon-a-chip systems [13–15]. This review presents recent advances in
in vitro tissue culture models by primarily emphasizing on organ-ona-chip platforms useful for studying the performance of drug delivery
nanotherapeutics. The current challenges in the development of
drug delivery systems are highlighted and the use of organ-on-chips
as a potential solution is discussed by presenting specific examples of
relevant proof-of-concept studies.
79
80
E
2
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
Recent innovations in microfluidic technologies can provide novel
synthesis and screening methods to address issues of scalability,
batch-to-batch variability, and poor predictability that limit the pace
of clinical translation of nanotherapeutics [16]. The integration of
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
239
240
3.1. Vascular platforms
R
B
C
E
F
U
D
N
C
O
R
A
242
243
244
245
246
247
248
249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
Nanoparticles introduced intravenously have to traverse through 272
the vascular endothelial barrier to reach the target tissue. It is thus im- 273
portant to study the interactions of these carriers with the vasculature 274
E
241
T
218
219
C
216
217
R
O
O
F
238
215
P
3. Organ-on-a-chip platforms as a potential solution
213
214
D
237
211
212
advanced 3D tissue engineered constructs with microfluidic network
systems, termed ‘organ-on-a-chip’, provides a novel platform for better
preclinical testing of drug delivery systems. Easy manipulation of
micro-liter volume of liquids has made these models a platform where
scaling and dynamic crosstalk between cells can be achieved [15]. The
system geometry and structures recreate physiological length scales,
concentration gradients, and the fluid flow generates mechanical
forces that recapitulate the in vivo microenvironment experienced by
cells [13,55]. Thus, these highly biomimetic platforms overcome the
drawbacks with conventional tissue culture models as highlighted
in the previous section [16]. By better mimicking the physiological
conditions and more accurately predicting the effect of drug carriers,
these sophisticated in vitro screening models can fill the gap between
the outcomes of animal studies and human clinical trials [16].
Since lung-on-a-chip [13,56], the field of biomimetic organs-on-achip has expanded rapidly to encompass several organs including
liver, kidney, heart, gut, breast, and blood vessels [55,57–59]. These
studies have already shown that organ-on-a-chip platforms can generate responses similar to those observed in vivo toward nanoparticles
[13,17]. Furthermore, multiple organ modules can be interconnected
in a physiologically relevant scale and organized to form a human-ona-chip platform [60]. These systems can be used to study the fate of
nanoparticles. The information obtained from such studies would
allow rapid testing of new therapeutic designs. Thus, the use of physiologically relevant organ-on-a-chip models as a studying tool for drug
delivery systems can accelerate the clinical translation of nanoparticles.
The following sections of this review, ordered by organ-type, will
discuss the progress, advantages and challenges of existing organ-ona-chip platforms, in the context of testing of drug delivery systems.
E
236
optimization studies. Evaluating the drug carrier in static in vitro assays
without considering mechanical forces and fluid flow has shown instances of false positives [13,17,22]. Hence, perfusion-based dynamic
culture systems in bioreactors have become increasingly popular. At
the same time, owing to the rapid development of microfluidics, the
size of these bioreactors is being constantly reduced while the functionality is improved, enabling the development of improved systems for
drug toxicity and efficacy studies [48–52]. For example, Lee and coworkers constructed microfluidic chambers containing arrays of hydrodynamic traps, which was used to immobilize around ten tumor cells
per trap (Fig. 2A). The entrapped tumor cells aggregated in the traps
to form small spheroids (b50 μm) within several hours (Fig. 2B, C)
[53]. Similarly, Cheung and co-workers developed a type of cell culture
chip with arrays of microsieves for efficient trapping of larger cell aggregates [54]. Treating the multicellular spheroids of LCC6/Her2 breast
tumor cells in the microsieves with increasing concentrations of DOX,
resulted in a reduction of the size of spheroid and viability of the cells.
In addition a microfluidic platform to study the interaction of nanoparticle–aptamer conjugates with prostate cancer cells that expressed
(LNCaP) or did not express (PC3) prostate specific surface antigen
(PSMA) was developed [11]. For LNCaP cells, the binding of nanoparticles was higher in the presence of aptamer and decreased with increasing flow rate, whereas in the case of PC3 the nanoparticles did not bind
with or without aptamers (Fig. 3). Such advanced microfluidic culture
models, where essentially any form of cell/tissue constructs (e.g., spheroids, hydrogels, and porous scaffolds) can be directly utilized on-chip
for testing, can potentially be used for cytotoxicity and efficacy assessment of drug delivery systems.
209
210
3
Q10 Fig. 1. The application of 3D scaffold-based multicellular tumor spheroids in drug testing. A–D) Confocal micrographs of live/dead staining showing (A) 2D culture and (C) 3D spheroid
culture. B) 2D culture revealed significant cell death after CdTe NP exposure. D) Spheroids showed much lower cell damage (especially in the central area) than the cells in 2D culture.
E, F) SEM images of (E) 2D culture and (F) 3D spheroid culture at 24 h post CdTe treatment. E) In 2D culture most cells were dead with a large amount of cells detached. F) Cells in spheroids
experienced much lower cellular damage than those in 2D culture.
Reproduced with permission from Ref. [39].
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
E
C
T
E
D
P
R
O
O
F
4
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
R
O
278
279
with regard to their vascular transport and toxicity. Some studies have
already reproduced geometric features of vasculature, including straight
channels [61–64], bifurcations [65,66], and a mixture of more complex
features [67]. By mapping and replicating a microvascular network
from a hamster muscle, a small microfluidic platform was created,
reproducing several geometric features including bifurcations and
tortuosities, as well as accounting for different shear stresses along
the branches [67]. Another important parameter to be considered is particle size. A microfluidic vascular model was developed to study the differences in accumulation of micro- and nanosized spheres inside
microchannels. It was reported that microspheres tend to locate more
to the margins when compared to nanospheres [68]. This effect of size
on accumulation, as well as hemodynamics and hemorheology, should
be taken into account when designing drug delivery carriers targeted
for vascular diseases and cancer. Additionally, these carriers also interact
with blood components. A study using high concentrations of mesoporous silica nanoparticles showed that despite having no effects on platelet
viability, it greatly increased platelet aggregation and adhesion to endothelium [62]. This emphasizes the evaluation of other parameters apart
from viability when studying particle interaction with blood components.
Another key aspect that vascular models are able to mimic is the
influence of shear stress [63,65,69]. This is particularly important for
nanoparticle delivery as it affects the endocytic uptake of nanoparticles
by endothelial cells [69]. Narrowing in particular regions of the vascular
system exhibits increased shear stress, which is an important factor in
C
276
277
U
N
275
R
Fig. 2. The application of microfluidic bioreactors for drug testing. A) Schematic showing the tumor spheroid formation and culture in a microfluidic flow chamber. B) Optical micrograph
showing that MCF-7 cells could uniformly fill all the traps. C) Optical micrographs showing the structure of the tumor spheroids: (left to right) phase contrast image and fluorescence
images showing cell membrane and nuclei.
the development of thrombosis. A microfluidic model was developed
to study the potential of shear responsive nanoparticles for targeting
and treating embolic occlusions. The tissue plasminogen activator
(tPA) loaded nanoparticles were able to dissolve preformed fibrin
clots inside this microfluidic channel. This in vitro observation was
then validated in an ex-vivo mouse pulmonary embolism model, a
shear-sensitive particle system allowed the use of a 100-fold lower
dose of tPA to obtain the same lysing effect as achieved by free tPA
(Fig. 4A) [63]. Such smart particles, which respond to biophysical cues,
can target drugs more efficiently, reduce dosage and concomitantly
lessen undesired secondary effects [63].
Vascular microfluidic models also take into account other aspects,
such as particle functionalization [61,65]. Platforms can model the influence of particle functionalization combined with either shear stress [61]
or geometric features as straight versus bifurcating channels [65]. Some
studies have highlighted the effect of particle shape on adhesion to the
channel walls (Fig. 4B) [66,70]. In case of an endothelialized channel,
rod-shaped nanoparticles showed high specific targeting and lower
non-specific accumulation as compared to sphere-shaped nanoparticles
[70]. When engineering and designing novel carriers, one should take
these shape effects into consideration.
Microfluidic technologies have the potential for scalability and highthroughput analysis, and can characterize populations of nanoparticles
in a quick and low cost manner [10], or even evaluate endothelial permeability [64]. Moreover, inherent characteristics of microfluidics,
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314
315
316
317
318
319
320
321
322
323
324
5
P
R
O
O
F
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
T
C
E
R
R
O
C
329
330
of action of drug carriers. A future challenge that still remains to be
elucidated is the prioritization of parameters, and, perhaps, how to
combine multiple features in a single, high-throughput capable, device.
On-chip vasculature models are thus a relevant tool when considering
the targeting of vascular diseases, tumor angiogenesis and usage of
injectable drug delivery systems.
N
327
328
such as laminar flow, can be exploited to build superior models.
For instance, hydrodynamic focusing [71] could be used for precisely
targeting drug carriers to specific regions in microfluidic chips, and
possibly study their subsequent transport to neighboring cells from a
non-exposed region. Several of the above discussed models have the
potential to leverage drug delivery by probing the efficacy and mode
U
325
326
E
D
Fig. 3. The application of microfluidic system for studying nanoparticle and cell interactions. A) Fluorescence images showing binding of rhodamine-labeled nanoparticles, without or with
PSMA aptamer, to PSMA-positive LNCaP cell and PSMA-negative PC3 cells under static conditions. The left column shows phase contrast images of the cells and corresponding fluorescence
images of the nanoparticles are indicated in the right column. B) Fluorescence images showing binding of rhodamine-labeled nanoparticle–aptamer conjugates (red) to LNCaP cells under
fluid flow conditions at flow rates of 0.25, 1, and 4 μL/min. The cells were stained with Calcein AM (green) and DAPI (blue) to quantify cell viability. C) Quantification of the number of
nanoparticles bound to PC3 cells under static condition and to LNCaP cells under both static and fluid flow conditions. (For interpretation of the references to color in this figure legend,
the reader is referred to the web version of this article.)
Reproduced with permission from Ref. [11].
Fig. 4. Microfluidic platforms provide insights on flow dynamics and shape influence for carrier attachment. A) Stenotic regions have higher shear rate (top) and accumulate more
nanoparticles (NP; bottom). B) Influence of particle shape (top) on attachment and accumulation in a 45° bifurcation, showing that disc-shaped particles attach more than spherical
particles (bottom).
Reproduced with permission from Ref. [63] and [66].
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
331
332
333
334
335
336
6
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
3.2. Cardiac platforms
3.3. Liver platforms
402
338
339
The need for targeting drugs for cardiovascular disease has become
very critical [72]. Several drugs used for cardiovascular diseases have
been subjected to FDA withdrawals [73]. Also a few commercial drugs
used to treat other organ disorders have been identified to have adverse
side effects on the cardiac tissue [74]. Inadequate screening models have
become one of the major accountable factors for these failures [74,75].
With multitude of features to be researched for a drug and its carrier
to successfully translate to clinics, testing them in the appropriate
in vitro conditions has turned out to be the key aspect [16]. However,
persisting difficulties in mimicking the physiology of the heart in vitro
have hampered this process. Nevertheless, there are several reports on
cardiac tissue engineering [76] and heart-on-a-chip [75] that promise
to potentially emerge as physiologically relevant models for in vitro
drug testing. Most of the developed platforms use primary cells derived
from rats, however, more focus on integration of human cells with these
models is required for mimicking human physiological response.
Among the human cell sources, cardiomyocytes generated from
human induced pluripotent stem cells have enormous potential to be
pathophysiologically relevant [77]. They also facilitate the creation of
platforms for mimicking genetic disorder in patient-specific cases,
thereby paving the way for individualized therapy [77].
The culture of functional cardiomyocytes can benefit from stimulation with electrical and mechanical forces [78], which cannot be
reproduced in a simple cell culture setting. Additionally, to recreate a
functional cardiac tissue, scaffolds can be used to recreate 3D tissue architecture [79]. The use of cardiomyocyte cell sheets has offered a possible platform for drug testing that exhibits an important physiological
parameter of heart: a uniform contractile function. Stevens et al. [80]
developed a scaffold free cardiac patch derived from human embryonic
stem cells (hESCs) that showed electromechanical coupling with
each beat. Shimizu et al. [81] fabricated these sheets with primary rat
myocytes. When two of these sheets were overlaid, they exhibited a
simultaneous and spontaneous beating after a week. Other attempts
to record electrical stimulation have involved the use of cardiac tissue
slices. Bussek et al. [82] implemented the use of guinea pig cardiac tissue
slices to evaluate the performance of a potassium channel blocker. Even
though different potentials were recorded when the slice was exposed
to the drug, the experiment does not completely reproduce the effects
in the human patient. Naturally, these reconstructions do not perform
as the organ itself and fail to reproduce in vivo conditions but they can
offer an advantage for further improvements by means of microfluidic
platforms.
Classically, most of the microfluidic platforms are fabricated using
poly(dimethylsiloxane) (PDMS). To tune the stiffness of the PDMS surface for cardiomyocyte culture the microfluidic channels can be coated
with a hydrogel of controllable matrix stiffness [14]. In a study by
Annabi et al. two types of hydrogels, methacrylated gelatin (GelMA)
and methacrylated tropoelastin (MeTro) were compared [14]. The cell
attachment and alignment were dependent on the type of hydrogel
used, whereas the beating was dependent on the stiffness of the matrix.
In another report, cardiomyocytes were seeded on thin PDMS films
patterned with fibronectin and cultured within a bioreactor [83]. The
high-throughput format enabled testing of different geometric patterns
of fibronectin and monitoring up to forty thin films in real-time in
a single bioreactor. As the calcium dynamics of the cardiomyocytes
are directly affected by disease conditions, Martewicz et al., developed a
microfluidic platform where the calcium dynamics of the cardiomyocytes
can be observed in real-time [84]. This platform could also control the
oxygen concentration experienced by the cells, thus allowing induction
of hypoxia. Concurrently, the differential response of calcium dynamics
was monitored with varying oxygen concentration. These in vitro
microfluidic platforms, with high-throughput capability, can accelerate
the translation of a drug carrier to the clinic by recreating cardiovascular
environment that better mimics human physiological conditions [75].
Drug hepatotoxicity is one of the main concerns in identifying new
drugs [85], thus studying hepatoxicity of nanoparticles is very important for developing novel drug delivery platforms. Also with animal
models failing to better predict outcomes of drug intake in humans
[85], there is an extensive body of research focused toward developing
in vitro models for evaluating hepatotoxity [86–89]. In vitro hepatic
models aim at emulating functionality and mimicking the normal as
well as pathophysiological architecture of liver. These models include
2D/3D mono- and co-cultures, hydrogel based engineered models, and
organ-on-a-chip platforms of normal and diseased cells. Since most of
the drug delivery and nanotoxicity studies are restricted to 2D cell
culture plates [90–92], some important studies that describe the testing
of nanoparticles in these platforms that more accurately predict in vivo
behavior are highlighted.
A recent study using lipid nanoparticles for siRNA delivery demonstrated the importance of using primary hepatocytes instead of immortalized cell lines for better translation of screening results to in vivo
outcomes [93]. Assessment of nanoparticles should be evaluated within
such clinically relevant models that can maintain the functionality of
primary hepatocytes for long-term culture [86,94]. One such model
used micropatterned collagen substrate for culturing primary hepatocytes in a high-throughput 24-well plate format [86]. In this model,
primary hepatocytes were co-cultured with fibroblasts to augment
homotypic interactions with heterotypic interactions for maintaining
the functionality of hepatocytes for a long period. This model represents
a potentially useful platform for long-term analysis of liver nanotoxicity.
In a study by Dragoni et al., the uptake and toxicity of AuNPs were investigated using precision cut rat liver slices [95]. The tissue slice model
was used to evaluate the uptake of AuNPs by hepatocytes, Kupffer and
endothelial cells present in the liver slice. However, tissue biopsies or
slices cannot be used for high-throughput studies and show a rapid
loss of functionality within days during in vitro culturing, thus limiting
their application for long-term studies [96].
With extensive research on developing 3D hepatic spheroids
for drug testing [87,88,97,98] and also with availability of highthroughput commercial platforms for formation of spheroids, liver
spheroids represent a promising 3D construct for rapid clinically relevant evaluation of nanoparticles. Spheroids formed from primary
human hepatocytes have been used for long-term drug studies [97]. In
a work by Lee et al., the toxicity of gold nanoparticles and CdTe nanoparticles in 2D cell cultures and 3D spheroids were compared (Fig. 1) [39].
Results showed considerably reduced toxicity in spheroids when compared to 2D cultures and the reduction in toxicity was attributed to
change in phenotype and complex cell–cell interaction that altered
the transport of nanoparticles. Spheroids of cancerous hepatocytes
have also been used as a model for mimicking in vivo tumor microenvironments as shown by England et al. [99]. They studied nanoparticle
delivery to avascular regions in the spheroid core and used surface
modification strategy to enhance nanoparticle penetration within the
tumor core.
Nanoparticles exhibit differential behavior in static and flow
conditions [13,17], thus necessitating the development of dynamic
microfluidic platforms for toxicity evaluation. Multiple biomimetic
liver-on-a-chip platforms have been long established for drug toxicity
testing [15,89,96,100]. Since liver is the major organ for drug metabolism, it is important to integrate a liver module with other organ
modules to assess the cytotoxicity of prodrugs. In such models, the
prodrugs are first metabolized by the liver module before reaching the
target organ of drug action. For example, Shuler and Sung developed a
micro cell culture analog (μCCA) with 3D cultures of HepG2/C3A cells
(as the liver module) and HCT-116 cells (as the tumor module), both
in Matrigel, and Kasumi-1 myeloblasts (as the bone marrow module)
in stiffer alginate gel (Fig. 5) [101]. The cytotoxic effect of Tegafur, an
oral prodrug of 5-fluorouracil (5-FU), on each organ analog was then
403
404
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398
399
400
401
O
R
O
P
359
360
D
357
358
E
355
356
T
353
354
C
351
352
E
349
350
R
347
348
R
345
346
O
343
344
C
342
U
N
340
341
F
337
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423
424
425
426
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
7
C
T
E
D
P
R
O
O
F
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
R
E
Fig. 5. The multi-chamber μCCA device. A) A PDMS μCCA device consisting of three chambers, which represented uterus, liver, and mammary tissues, respectively. B) Measurement of cell
viability after treatment with 0.02% Triton. Triton treatment led to full mortality for all cell types.
Reproduced with permission from Ref. [101].
482
3.4. Lung platforms
483
484
The respiratory tract is one of the most significant ports of entry in
the human body. This can result in a number of infectious diseases as
well as occupational pathology, but it also offers an appealing route of
drug administration. The alveoli themselves offer a thin mucosal barrier
(200–800 nm) with low enzymatic action, vast absorptive surface area
(100 m2) and rapid access to the bloodstream while avoiding invasive
procedures [104]. Aerosol nanocomplexes for gene therapy have proven
to diffuse through the alveoli–capillary barrier in an efficient manner
compared to other particles due to their small size [105]. Other drug
476
477
478
479
485
486
Q3
487
488
489
490
491
O
474
475
C
472
473
N
470
471
U
468
469
R
480
481
tested using such a three organ-on-a-chip platform, where they found
that the μCCA was capable of reproducing the metabolism of Tegafur
to 5-FU in the liver module, whereas the metabolic conversion of
Tegafur could not be accomplished with 2D cultures in 96-well plates.
In a work by Wagner et al., a multi organ-on-a-chip model was developed for long-term culture by combining liver microtissues made
of spheroids of HepaRG and hepatic stellate cells with skin biopsies
[15]. Their microfluidic system integrated an inbuilt peristaltic pump
with a total media volume of 300 μL, thus enabling cross-talk between
skin and liver compartments proven by the dynamic level of albumin
in mono- and co-culture systems. Such models could be channeled
toward evaluating the toxicity of nanoparticles used in the cosmetic
industry. Infectious disease models that recreate the hepatic stage of infections, such as hepatitis C viral infection, will also prove significantly
important for clinical translation of nanotherapeutics [102,86,103].
467
delivery systems like zinc oxide nanoparticles (ZnONP) [106], silver
nanoparticles (AgNP) [107] and doxorubicin micelles [108] have been
used with 2D culture platforms to study the effect on cell function and
apoptotic response. Multi-well plate cultures of MCF-7 and A549 lung
cancer cell lines have been used to evaluate nanoparticles [109]. But
these systems fail to reproduce a physiological environment for drug testing. To recreate the complexity of the human lung's structure, the use of
3D microfluidic systems that can introduce fluid and solid mechanical
forces [110] is essential to imitate the human alveoli environment.
In an effort to develop biomimetic ‘lung-like’ microsystems, Huh
et al. reproduced the alveolar–capillary interface in vitro by creating a
layer of alveolar epithelial cells and endothelial cells both seeded on
opposite sides of a porous ECM-coated membrane (Fig. 6A) [13]. The
inflammatory and toxic effect of silica nanoparticles was evaluated by
particle absorption through a vacuum strain model that mimics the
physiological breathing motion. This cyclic mechanical force augmented
the nanoparticle translocation across the alveolar–capillary barrier, enhancing the toxic and inflammatory response as indicated by increased
reactive oxygen species (ROS) generation and intercellular adhesion
molecule-1 (ICAM-1) expression. The nanoparticle translocation behavior was also evaluated in a whole mouse lung ventilation–perfusion
model to demonstrate the significance of using lung-on-a-chip systems
to better predict physiological response. Yu et al. fabricated a ‘lung-likecompartment’ in one of the four multi-channel 3D microfluidic cell
culture platform that also incorporated kidney, liver and adipose cells
[111]. They evaluated gelatin microspheres as a controlled-release system for TGF-β1 encapsulated within the microspheres, and used A549
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
509
510
511
512
513
514
515
516
517
518
8
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
A
Alveolar
epithelium
C
B
Vacuum
compartment
Nanoparticles
Cells
Tubular cells
Vacuum
regulator
Porous membrane
Endothelium
ECM-coated
membrane
Lower layer
Interstitium
Cell chamber
Stretched
F
Unstretched
Other organs that are important for studying nanocarrier mediated
drug delivery are the kidney, spleen and gut [113–115]. Kidney and
spleen play an important role in the clearance of systemically delivered
nanoparticles. The kidney is one of the major excretion pathways in
the human body, playing a key role in blood filtration and water metabolism [116]. A study indicated that the glomerular filtration barrier contributed toward the dissociation of siRNA from its cationic cyclodextrin
carrier. Thus it is important to study the behavior of nanoparticles in the
renal system especially when administered intravenously [113]. To replicate renal conditions, such as fluid flow and shear stress [117], Jang
et al. created a microfluidic device that mimics the in vivo renal tubular
environment [118]. The device consists of a PDMS platform designed to
contain both ‘luminal’ and ‘tubular’ chambers separated by a porous
membrane. Human primary kidney proximal tubular epithelial cells
were grown on the ‘tubular’ side of the membrane while media was
perfused into the ‘luminal’ side (Fig. 6B) [118]. The device demonstrated
differential cell histologic arrangement and protein expression when
using static and dynamic flow conditions. The dynamic condition
revealed an augmentation in the reabsorption function of the kidney.
In the work by Shintu et al., a microfluidic device was used to measure
the ‘metabolomic’ fingerprint of the kidney using high-throughput
screening approach [119]. This method provides for rapid screening of
a compound target library that can be of further use in the drug delivery
and discovery research.
Spleen plays an important role in filtering the bloodstream and is a
vital part of the body's immunological response [120]. It forms a fundamental defense component against certain bacteria such as Streptococcus
pneumoniae. Yung et al. [121] created a blood cleansing a platform that
uses magnetic opsonins to target Candida albicans [121]. The pathogen
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
C
E
R
538
539
R
536
537
O
530
531
C
528
529
U
N
526
527
3.6. Tumor platforms
579
The complex in vivo microenvironment of a tumor presents a unique
challenge for the efficient delivery of nanotherapeutics to the tumor
site. The key features of this complex microenvironment include heterogenous vascularization, increased interstitial pressure and inadequate
lymphatic drainage [122]. Significant advances have been made in the
chemical discovery, design and synthesis of chemotherapeutic drugs
and diagnostic agents. However, their adverse side effects in healthy
tissues greatly limit the maximum tolerated dose and thereby reduce
their therapeutic efficacy [123–125]. Researchers and pharmaceutical
companies are pursuing the use of nanoparticles for encapsulating
anti-cancer drugs and delivering them specifically to the tumor site as
a potential solution to reduce the side effects [126]. Nanoparticles
with surface targeting moieties have shown efficacy in in vitro screening
studies, but fail to show the same performance during in vivo trials.
Several delivery barriers make nanotherapeutics ineffective within the
human body and hinder successful clinical translation of these anticancer drug carriers. To solve this delivery problem, it is important to
gain an understanding of the nanoparticle transport through the bloodstream, distribution at the target tissue and subsequent uptake by the
cells. Preclinical models that mimic the in vivo 3D tumor architecture
and dynamic flow conditions are important for studying these delivery
parameters and for toxicology assessment.
The integration of microfluidic networks with 3D tissue engineered
cultures offers a unique opportunity to probe nanoparticle transport
barriers in a controlled manner. Additionally, these tumor-on-a-chip
platforms can be used for studies to optimize nanoparticle dosage, to
generate gradient drug concentrations and to develop personalized
580
581
P
534
535
524
525
563
D
3.5. Other organ platforms
522
523
is then removed by a micromagnetic–microfluidic separator and cleared
from the blood. Such models are yet to be exploited for drug studies and
can be of great use for optimizing drug delivery systems.
Among other organs that could be used for drug testing is the guton-a-chip created by Kim et al. [55] This device recreates intestinal
human environment and could be used to test oral drug absorption.
The device consists of two chambers divided by a porous membrane
(Fig. 6C). The membrane separates a columnar epithelium of Caco-2
cells from another chamber that lies below the porous structure. In addition, Lactobacillus rhamnosus GG bacteria was cultured together with
the epithelial cells to enhance the barrier function of cells during this
period. The inclusion of a Lactobacillus that resembles the intestinal
flora offers a step forward toward simulating the intestinal lumen.
Future design strategies ought to consider the development of an
organ-like platform that can mimic the in vivo complexity of the gut
environment in a simple and efficient way.
E
533
520
521
T
532
cells to recreate a human lung microenvironment for drug testing. In the
device, common media goes through this first chamber of A549 cells
and TGF-β1 encapsulated microspheres to continue to the other ‘organs’. The ‘lung’ compartment revealed an enhanced cellular function
compared to the other uncompromised ‘organs’ showing a compartmental separation between the chambers and imitating the partial
cross-talk between organs that occurs in the human body.
Future endeavors must be oriented to combine the multiple cell
types in the alveoli and the mechanical forces involved in ventilation.
The reconstruction of a reliable alveolar–capillary barrier without a
complex culturing process [112] as well as developing novel particles
with effective transport properties are also upcoming challenges. A
system that combines all of the above would be a reliable model that
represents the inner organ conditions in the human body.
519
R
O
O
Fig. 6. A) Schematic of the lung-on-a-chip platform developed to study nanoparticle transport. The transport of nanoparticles across the alveolar–capillary interface is augmented significantly when vacuum is applied to the side channels of the lung platform. Adapted from Ref. [13]. B) Schematic of the kidney-on-chip platform with two chambers divided by a porous
membrane that separates the ‘luminal’ from the ‘tubular’ space recreating the in vivo conditions in the kidney. Adapted from Ref. [118]. C) The gut-on-a-chip platform includes two compartments divided by a porous membrane separating Caco-2 cells from the bottom chamber. A vacuum regulator is attached to the side chambers to assess the behavior of the cells under
the effect of strain. Adapted from Ref. [55].
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
564
565
566
567
568
569
570
571
572
573
574
575
576
577
578
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
R
O
O
F
630
631
P
628
629
D
626
627
E
624
625
T
622
623
C
620
621
E
618
619
R
616
617
R
614
615
O
613
C
611
612
different concentrations and combinations of anti-cancer drugs on a parallel culture of PC3 prostate cancer cells. Their platform could capture the
synergy between different sensitizer drugs (DOX or mitoxantrone in this
case) and TRAIL (TNF-alpha Related Apoptosis Inducing Ligand). Such
high-throughput systems can be used for screening and optimizing combinatorial delivery strategies using nanocarriers for cancer therapeutics
before testing in humans [135,136]. Further, microfluidic systems that
allow for testing anti-cancer drug sensitivity to a specific patient are
promising for developing clinically relevant nanotherapeutics for personalized treatments [137]. The study by Zhao et al. addressed a key aspect of anti-cancer drug assessment, namely the integration of
quantitative data acquisition methods with the on-chip platform for analyzing the cell apoptosis induced by the chemotherapeutic drug. This
was achieved by using Annexin V conjugated quantum dots and Calcein
AM as dual apoptotic probes [138]. Such real-time optical probing
methods will be useful for monitoring the behavior of nanotherapeutics
in the tumor-on-a-chip platforms.
New delivery systems have been proposed in recent years to
meet the demand for developing safer chemotherapeutic strategies for
breast cancer, one of the commonest types of tumor in women [139].
These include liposomal nanoparticles used to deliver DOX [140] and
albumin-bound Paclitaxel delivery system [141]. The anatomy of the
mammary gland presents a number of challenges when it comes to clinical translation of these theranostics. Two of the major barriers are: the
presence of a system of ducts in a branched manner with different sizes
N
609
610
drug screening and treatment strategies [127]. For example, in a recent
report by Albanese et al., a tumor-on-a-chip model was developed to
investigate the transport behavior of AuNPs through 3D tumor-like
spheroids of human melanoma cells immobilized in a PDMS chamber
(Fig. 7A) [128]. Their microfluidic device allowed precise control of
the media flow conditions and the spheroids were coated with a laminin layer that acted as a barrier to AuNP transport, thus mimicking
conditions found in vivo at the tumor site. The tissue penetration and
accumulation of fluorescent NPs under physiological flow conditions
were monitored in real-time. The nanoparticle diameter, surface
functionalization and the flow conditions in the microenvironment
were shown to affect the accumulation of AuNPs near the tumor tissue
(Fig. 7B, C). The nanoparticles functionalized with targeting groups
accumulated in the periphery and failed to penetrate deep inside the
core. Such studies provide critical insights to help design better nanoparticles for improved in vivo targeting. Pellegrino et al. have also designed a microfluidic platform to evaluate magnetic thermoresponsive
polymers for controlled release of chemotherapeuitc drugs. This
preliminary study was conducted without cells in PBS, but this system
can be further developed for studying drug delivery to cells using
thermoresponsive carriers [129].
Several other tumor-on-a-chip platforms have been reported
to study the complex tumor microenvironment and to screen drugs
[127,130–135]. In a recent study by Kim et al., a fully automated
microfluidic system was developed for high-throughput screening of
U
607
608
9
Fig. 7. Studying the nanoparticle (NP) tissue transport behavior using tumor-on-chip platform. A) The schematic of the microfluidic chip assembled on top of a microscope stage. B) The
schematic (left), image (center) and graph (right) showing the effect of NP size on tissue accumulation. Four different sizes, 40 (red), 70 (blue), 110 (green) and 150 (orange) of PEGylated
NPs were investigated. C–D) The effect of NP functionalization on tissue accumulation: C) PEGylated NPs and D) iron-transporting transferring (Tf) protein functionalized NP.
(For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
Reproduced with permission from Ref. [128].
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
681
682
Pharmaceutical companies involved in drug discovery continuously
aim to both reduce the cost of their research and speed up the development of new drugs. In this process, pre-clinical animal studies are the
most expensive and time-consuming part, while not assuring human
in vivo significance. In vitro organ-on-a-chip platforms can revolutionize
this process by making it less expensive, while representing a reliable
way for drug discovery. However, further advances in the field are needed to overcome engineering and biological challenges before entirely
assessing their value as an efficient and robust platform for screening
nanotherapeutics.
The final goal is to develop a body-on-a-chip platform for systemic
evaluation of drug delivery vectors. But current efforts are directed
toward the development of individual organ platforms limiting their
range of applications to a specific organ's functions. For example, liver
being the most relevant organ for drug toxicity and metabolism, the development of a functional liver-on-a-chip system is the most important
challenge currently faced by researchers. As mentioned in Section 3.3,
several attempts have been developed in this direction, but the available
platforms are yet unable to fully capture the complexities of in vivo drug
metabolism in a robust manner. For developing a universal platform,
the flexibility and modularity of this technology, based on extremely
versatile microfabrication techniques, should be exploited. The possibility of culturing different cell types employing the identical standard
setups could lead to the development of several organ models on
demand. The results obtained with such systems could be easily compared, and relevant drug-specific cell responses can be quickly noticed.
In this context, the simplicity of the setup and the implementation of
several parallel experiments will be a key aspect (e.g., for testing
drug–dose responses as well as different target organs). Indeed, in
many cases the platforms proposed so far have been optimized for
one or two main parameters and the comparison between them is
highly complicated. A crucial challenge is to build platforms that deliver
comparable results, both to other systems and among different research
groups.
Furthermore, the inherent modularity of these systems allows for
the development of several on-chip modules by only changing cell
type. Interconnection of such modules would mimic the communication between different organs, making the search for potential new
drugs a more effective and comprehensive process. However, highthroughput and automation, which are intrinsic capabilities of these
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
Acknowledgments
781
D
E
683
684
T
676
677
C
674
675
E
672
673
R
670
671
R
668
669
O
666
667
C
664
665
U
N
663
F
4. Future directions & conclusion
661
662
721
722
O
680
659
660
systems, yet remain a challenge. Developments and efforts on the field
of biosensors should be coupled to organs-on-a-chip, so that the evaluation of outcomes could be done in a quantitative and real-time manner.
An ideal platform should include these sensors to monitor, in situ,
various physical/chemical parameters, such as O2, pH or even proteins.
A robust system has to show stable experimental conditions in order
to correlate any significant change in those parameters to the direct
effect of the candidate drug. Commercially available sensors, and
microfabricated ones, that allow a quick integration in a microfluidic
system, will be combined to previously developed organ-on-a-chip
platforms.
Future platforms should integrate parenchymal and nonparenchymal cells, and allow long-term experiments without
experiencing irreversible changes in cell function and behavior. Parameters such as cell proliferation and viability are quite immediate to assess,
but changes in proteomic, genomic and epigenomic levels, that can
dramatically affect drug metabolism, can be more difficult to monitor. A
stable and robust system to address these issues is a mandatory challenge
that will be faced in the next generation organ-on-a-chip systems.
New organ-on-a-chip platforms for drug discovery should also consider any problems arising from the interaction between a candidate
drug vector and the microfluidic systems. Mathematical and computational fluid dynamics (CFD) studies can help in this direction, providing
a mechanistic understanding of drug carrier attachment in organ-on-achip systems. The known differential behavior of nanoparticle shape in
terms of cell adhesion and distribution can be assessed by CFD, and
potentially aid in the generation of better and more comprehensive
models. For instance, in a computational study nanorods were shown
to contact and adhere to the wall much better than spherical counterparts [147]. Moreover, a CFD model of a branched blood vessel showed
that bifurcating sections had higher nanoparticle accumulation when
compared to straight ones. In the same study, authors were able to
model the trajectory and adhesive forces of nanoparticles under shear
flow [148]. Together, these and other works [149], set the stage for future CFD studies on particle–particle, particle–cell and particle–
microfluidic device interaction (accumulation and adhesion). More
complex CFD studies on these phenomena, especially within organon-a-chip, can help building next generation drug delivery systems, by
better understanding their underlying biophysical aspects.
From a material point of view, the devices developed in this field
show important differences that can determine different cell responses
as well as affect drug concentration into the system, due to drug nonspecific adsorption to the channel walls. Though PDMS and polystyrene
are the most popular materials due to several advantages [150], such as
optical transparency and cell compatibility, other plastic materials have
been used and a detailed comparison of their advantages/drawbacks in
drug screening tests is missing. If an ideal material will be not found,
research groups could decide to cover the microchannels with a cell
layer for preventing drug adsorption, as successfully reported by
Schimek et al. [151].
In the long run, organ-on-a-chip systems will be used also for
personalized drug screening. As these systems will show strong standardization and reliability, the possibility to use patient-derived iPSCs,
produced in vitro and then terminally differentiated, can solve one of
the most important problems in disease treatment — patient-to-patient
drug response variability — avoiding long and ineffective drug treatments and minimizing drug toxicity. In summary, organ-on-a-chip
platforms for studying drug delivery systems hold great promise and
some aspects of them have been investigated, but their full applicability
and potential have yet to be realized.
P
678
679
[142] and high pressure buildup during a diagnostic liquid procedure
like the fluid nipple aspiration or ductal lavage. Due to the increased
pressure in the smaller ducts, the delivered theranostics fail to reach
their target and in 20% of the patients these methods provide an inadequate number of cells to determine a diagnosis [143]. In their effort to
reproduce the ductal system in a mammary gland and address the difficulties mentioned above, Grafton et al. developed a microfluidic breaston-a-chip system. This system consisted of laminin coated microfluidic
channels with decreasing ductal size to culture a polarized monolayer of
human mammary epithelial cells. In this model, they proposed the use
of iron oxide superparamagnetic sub-micron particles (SMPs) with an
external magnetic field guidance for theranostic purposes of mammary
gland neoplasias [144]. To further investigate breast neoplasia models,
the same group attempted the reconstruction of tumor growth in the
mammary gland. Knowing that most of the human mammary lesions
ascend from the terminal lobular ductal units [145], they used a platform to reproduce the disease on a chip. Human breast epithelial cells
and nodules (3–5 cells) of tumorigenic human mammary carcinoma
cells were seeded in an acrylic hemichannel system leading to a
model that mimics the tumor's organization in human biopsies [146].
Using patient biopsies in future studies, the tumor-on-a-chip models
described above could serve as a personalized medicine platform to
optimize an individualized chemotherapy regimen.
657
658
R
O
10
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
771
772
773
774
775
776
777
778
779
780
The content is solely the responsibility of the authors and does not 782
necessarily represent the official views of the awarding agency. The 783
authors acknowledge funding from the Office of Naval Research 784
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
N
C
O
R
R
E
C
R
O
O
F
T
[1] O.C. Farokhzad, R. Langer, Nanomedicine: developing smarter therapeutic and
diagnostic modalities, Adv. Drug Deliv. Rev. 58 (2006) 1456–1459.
[2] N.A. Peppas, Intelligent therapeutics: biomimetic systems and nanotechnology in
drug delivery, Adv. Drug Deliv. Rev. 56 (2004) 1529–1531.
[3] G. Gregoriadis, Drug entrapment in liposomes, FEBS Lett. 36 (1973) 292–296.
[4] G. Gregoriadis, R.A. Buckland, Enzyme-containing liposomes alleviate a model for
storage disease, Nature 244 (1973) 170–172.
[5] D.L. Stirland, J.W. Nichols, S. Miura, Y.H. Bae, Mind the gap: a survey of how cancer
drug carriers are susceptible to the gap between research and practice, J. Control.
Release 172 (2013) 1045–1064.
[6] Q. Fu, J. Sun, W. Zhang, X. Sui, Z. Yan, Z. He, Nanoparticle albumin-bound (NAB)
technology is a promising method for anti-cancer drug delivery, Recent Pat.
Anticancer Drug Discov. 4 (2009) 262–272.
[7] N. Hakimi, S.S. Tsai, C.H. Cheng, D.K. Hwang, One-step two-dimensional
microfluidics-based synthesis of three-dimensional particles, Adv. Mater. (2013).
[8] P.M. Valencia, E.M. Pridgen, M. Rhee, R. Langer, O.C. Farokhzad, R. Karnik,
Microfluidic platform for combinatorial synthesis and optimization of targeted
nanoparticles for cancer therapy, ACS Nano 7 (2013) 10671–10680.
[9] J. Andreas, E.R. Joseph, N.V. Wyatt, L.D. Don, E.L. Laurie, G. Michael, Preparation of
nanoparticles by continuous-flow microfluidics, J. Nanoparticle Res. (2008).
[10] J.L. Fraikin, T. Teesalu, C.M. McKenney, E. Ruoslahti, A.N. Cleland, A highthroughput label-free nanoparticle analyser, Nat. Nanotechnol. 6 (2011) 308–313.
[11] O.C. Farokhzad, A. Khademhosseini, S. Jon, A. Hermmann, J. Cheng, C. Chin, A. Kiselyuk,
B. Teply, G. Eng, R. Langer, Microfluidic system for studying the interaction of nanoparticles and microparticles with cells, Anal. Chem. 77 (2005) 5453–5459.
[12] G.M. Whitesides, The origins and the future of microfluidics, Nature 442 (2006)
368–373.
[13] D. Huh, B.D. Matthews, A. Mammoto, M. Montoya-Zavala, H.Y. Hsin, D.E. Ingber,
Reconstituting organ-level lung functions on a chip, Science 328 (2010)
1662–1668.
[14] N. Annabi, S. Selimovic, J.P. Acevedo Cox, J. Ribas, M. Afshar Bakooshli, D. Heintze,
A.S. Weiss, D. Cropek, A. Khademhosseini, Hydrogel-coated microfluidic channels
for cardiomyocyte culture, Lab Chip 13 (2013) 3569–3577.
[15] I. Wagner, E.M. Materne, S. Brincker, U. Sussbier, C. Fradrich, M. Busek, F. Sonntag,
D.A. Sakharov, E.V. Trushkin, A.G. Tonevitsky, R. Lauster, U. Marx, A dynamic multiorgan-chip for long-term cultivation and substance testing proven by 3D human
liver and skin tissue co-culture, Lab Chip 13 (2013) 3538–3547.
[16] P.M. Valencia, O.C. Farokhzad, R. Karnik, R. Langer, Microfluidic technologies for accelerating the clinical translation of nanoparticles, Nat. Nanotechnol. 7 (2012) 623–629.
[17] S.K. Mahto, T.H. Yoon, S.W. Rhee, A new perspective on in vitro assessment
method for evaluating quantum dot toxicity by using microfluidics technology,
Biomicrofluidics 4 (2010).
[18] W. Jiang, Y.S. KimBetty, J.T. Rutka, W.C.W. Chan, Nanoparticle-mediated cellular
response is size-dependent, Nat. Nanotechnol. 3 (2008) 145–150.
[19] B.D. Chithrani, A.A. Ghazani, W.C.W. Chan, Determining the size and shape
dependence of gold nanoparticle uptake into mammalian cells, Nano Lett. 6
(2006) 662–668.
[20] J. Baselga, L. Norton, J. Albanell, Y.-M. Kim, J. Mendelsohn, Recombinant humanized
anti-HER2 antibody (Herceptin(TM)) enhances the antitumor activity of Paclitaxel
and Doxorubicin against HER2/neu overexpressing human breast cancer xenografts, Cancer Res. 58 (1998) 2825–2831.
[21] A. Monks, D. Scudiero, P. Skehan, R. Shoemaker, K. Paull, D. Vistica, C. Hose, J.
Langley, P. Cronise, A. Vaigro-Wolff, M. Gray-Goodrich, H. Campbell, J. Mayo, M.
Boyd, Feasibility of a high-flux anticancer drug screen using a diverse panel of
cultured human tumor cell lines, J. Natl. Cancer Inst. 83 (1991) 757–766.
[22] E.C. Cho, Q. Zhang, Y. Xia, The effect of sedimentation and diffusion on cellular
uptake of gold nanoparticles, Nat. Nanotechnol. 6 (2011) 385–391.
[23] N.S. Bhise, R.S. Gray, J.C. Sunshine, S. Htet, A.J. Ewald, J.J. Green, The relationship between terminal functionalization and molecular weight of a gene delivery polymer
and transfection efficacy in mammary epithelial 2-D cultures and 3-D organotypic
cultures, Biomaterials 31 (2010) 8088–8096.
[24] S. Wilkening, F. Stahl, A. Bader, Comparison of primary human hepatocytes and
hepatoma cell line Hepg2 with regard to their biotransformation properties,
Drug Metab. Dispos. 31 (2003) 1035–1042.
[25] G. Hamilton, Multicellular spheroids as an in vitro tumor model, Cancer Lett. 131
(1998) 29–34.
[26] S.-M. Ong, Z. Zhao, T. Arooz, D. Zhao, S. Zhang, T. Du, M. Wasser, D. van Noort, H. Yu,
Engineering a scaffold-free 3D tumor model for biN in vitrob/iN drug penetration
studies, Biomaterials 31 (2010) 1180–1190.
[27] C. Dubessy, J.-L. Merlin, C. Marchal, F. Guillemin, Spheroids in radiobiology and
photodynamic therapy, Crit. Rev. Oncol. Hematol. 36 (2000) 179–192.
[28] L.A. Kunz-Schughart, Multicellular tumor spheroids: intermediates between
monolayer culture and in vivo tumor, Cell Biol. Int. 23 (1999) 157–161.
[29] W. Mueller-Klieser, Multicellular spheroids, J. Cancer Res. Clin. Oncol. 113 (1987)
101–122.
U
791
792
793
794
795
796
797
798
799
800
801
802
803
804
805
Q5
806
807
808
809
810
Q6
811
812
813
814
815
816
817
818
819
820
821
822
823
824
825
826
827
828
829
830
831
832
833
834
835
836
837
838
839
840
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
P
References
D
790
787
788
[30] J.M. Yuhas, A.P. Li, A.O. Martinez, A.J. Ladman, A simplified method for production
and growth of multicellular tumor spheroids, Cancer Res. 37 (1977) 3639–3643.
[31] Y.-C. Tung, A.Y. Hsiao, S.G. Allen, Y.-s. Torisawa, M. Ho, S. Takayama, Highthroughput 3D spheroid culture and drug testing using a 384 hanging drop
array, Analyst 136 (2011) 473–478.
[32] J.M. Karp, J. Yeh, G. Eng, J. Fukuda, J. Blumling, K.-Y. Suh, J. Cheng, A. Mahdavi, J.
Borenstein, R. Langer, A. Khademhosseini, Controlling size, shape and homogeneity
of embryoid bodies using poly(ethylene glycol) microwells, Lab Chip 7 (2007)
786–794.
[33] H. Hardelauf, J.-P. Frimat, J.D. Stewart, W. Schormann, Y.-Y. Chiang, P. Lampen, J.
Franzke, J.G. Hengstler, C. Cadenas, L.A. Kunz-Schughart, J. West, Microarrays for
the scalable production of metabolically relevant tumour spheroids: a tool for
modulating chemosensitivity traits, Lab Chip 11 (2011) 419–428.
[34] M.A. Kinney, R. Saeed, T.C. McDevitt, Systematic analysis of embryonic stem cell
differentiation in hydrodynamic environments with controlled embryoid body
size, Integr. Biol. 4 (2012) 641–650.
[35] Y. Zhang, Y. Xia, Formation of embryoid bodies with controlled sizes and maintained pluripotency in three-dimensional inverse opal scaffolds, Adv. Funct.
Mater. 22 (2012) 121–129.
[36] Y.S. Torisawa, A. Takagi, Y. Nashimoto, T. Yasukawa, H. Shiku, T. Matsue, A multicellular spheroid array to realize spheroid formation, culture, and viability assay on a
chip, Biomaterials 28 (2007) 559–566.
[37] Y.S. Zhang, S.W. Choi, Y.N. Xia, Inverse opal scaffolds for applications in regenerative medicine, Soft Matter 9 (2013) 9747–9754.
[38] B. Kim, G. Han, B.J. Toley, C.-k. Kim, V.M. Rotello, N.S. Forbes, Tuning payload
delivery in tumour cylindroids using gold nanoparticles, Nat. Nanotechnol. 5
(2010) 465–472.
[39] J. Lee, G.D. Lilly, R.C. Doty, P. Podsiadlo, N.A. Kotov, In vitro toxicity testing of
nanoparticles in 3D cell culture, Small 5 (2009) 1213–1221.
[40] L.A. Gurski, A.K. Jha, C. Zhang, X. Jia, M.C. Farach-Carson, Hyaluronic acid-based
hydrogels as 3D matrices for in vitro evaluation of chemotherapeutic drugs using
poorly adherent prostate cancer cells, Biomaterials 30 (2009) 6076–6085.
[41] F.M. Kievit, S.J. Florczyk, M.C. Leung, O. Veiseh, J.O. Park, M.L. Disis, M. Zhang,
Chitosan-alginate 3D scaffolds as a mimic of the glioma tumor microenvironment,
Biomaterials 31 (2010) 5903–5910.
[42] D.W. Hutmacher, Biomaterials offer cancer research the third dimension, Nat.
Mater. 9 (2010) 90–93.
[43] A.J. Engler, S. Sen, H.L. Sweeney, D.E. Discher, Matrix elasticity directs stem cell
lineage specification, 126 (2006) 677–689.
[44] D.E. Discher, P. Janmey, Y.-l. Wang, Tissue cells feel and respond to the stiffness of
their substrate, Science 310 (2005) 1139–1143.
[45] N. Huebsch, P.R. Arany, A.S. Mao, D. Shvartsman, O.A. Ali, S.A. Bencherif, J. RiveraFeliciano, D.J. Mooney, Harnessing traction-mediated manipulation of the cell/matrix
interface to control stem-cell fate, Nat. Mater. 9 (2010) 518–526.
[46] J. Liu, Y. Tan, H. Zhang, Y. Zhang, P. Xu, J. Chen, Y.-C. Poh, K. Tang, N. Wang, B.
Huang, Soft fibrin gels promote selection and growth of tumorigenic cells, Nat.
Mater. 11 (2012) 734–741.
[47] C. Fischbach, R. Chen, T. Matsumoto, T. Schmelzle, J.S. Brugge, P.J. Polverini, D.J.
Mooney, Engineering tumors with 3D scaffolds, Nat. Methods 4 (2007) 855–860.
[48] M.-H. Wu, S.-B. Huang, G.-B. Lee, Microfluidic cell culture systems for drug
research, Lab Chip 10 (2010) 939–956.
[49] M.-H. Wu, S.-B. Huang, Z. Cui, Z. Cui, G.-B. Lee, Development of perfusion-based
micro 3-D cell culture platform and its application for high throughput drug testing, Sens. Actuators B 129 (2008) 231–240.
[50] R. Baudoin, L. Griscom, J.M. Prot, C. Legallais, E. Leclerc, Behavior of HepG2/C3A cell
cultures in a microfluidic bioreactor, Biochem. Eng. J. 53 (2011) 172–181.
[51] S.T. Yang, X. Zhang, Y. Wen, Microbioreactors for high-throughput cytotoxicity
assays, Curr. Opin. Drug Discov. Devel. 11 (2008) 111–127.
[52] C. Robert, E. Kamal, W. Honglu, S. Wei, Biofabrication of a three-dimensional liver
micro-organ as an in vitro drug metabolism model, Biofabrication 2 (2010)
045004.
[53] L.Y. Wu, D. Di Carlo, L.P. Lee, Microfluidic self-assembly of tumor spheroids for
anticancer drug discovery, Biomed. Microdevices 10 (2008) 197–202.
[54] L. Yu, M.C. Chen, K.C. Cheung, Droplet-based microfluidic system for multicellular
tumor spheroid formation and anticancer drug testing, Lab Chip 10 (2010)
2424–2432.
[55] H.J. Kim, D. Huh, G. Hamilton, D.E. Ingber, Human gut-on-a-chip inhabited by
microbial flora that experiences intestinal peristalsis-like motions and flow, Lab
Chip 12 (2012) 2165–2174.
[56] D. Huh, H. Fujioka, Y.C. Tung, N. Futai, R. Paine 3rd, J.B. Grotberg, S. Takayama,
Acoustically detectable cellular-level lung injury induced by fluid mechanical
stresses in microfluidic airway systems, Proc. Natl. Acad. Sci. U. S. A. 104 (2007)
18886–18891.
[57] J. Sung, M. Shuler, In vitro microscale systems for systematic drug toxicity study,
Bioprocess Biosyst. Eng. 33 (2010) 5–19.
[58] A. Sin, K.C. Chin, M.F. Jamil, Y. Kostov, G. Rao, M.L. Shuler, The design and fabrication of three-chamber microscale cell culture analog devices with integrated
dissolved oxygen sensors, Biotechnol. Prog. 20 (2004) 338–345.
[59] D. Huh, G.A. Hamilton, D.E. Ingber, From 3D cell culture to organs-on-chips, Trends
Cell Biol. 21 (2011) 745–754.
[60] C. Moraes, J.M. Labuz, B.M. Leung, M. Inoue, T.H. Chun, S. Takayama, On being
the right size: scaling effects in designing a human-on-a-chip, Integr. Biol.
(Camb.) 5 (2013) 1149–1161.
[61] J. Kusunose, H. Zhang, M.K. Gagnon, T. Pan, S.I. Simon, K.W. Ferrara, Microfluidic
system for facilitated quantification of nanoparticle accumulation to cells under
laminar flow, Ann. Biomed. Eng. 41 (2013) 89–99.
E
789
Young National Investigator Award, the National Institutes of Health
(HL092836, DE019024, EB012597, AR057837, DE021468, HL099073,
EB008392), and the Presidential Early Career Award for Scientists and
Engineers (PECASE). J.R. acknowledges the support from the Portuguese
Foundation for Science and Technology (FCT; SFRH/BD/51679/2011).
785
Q4
786
11
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
866
867
868
869
870
871
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
901
902
903
904
905
906
907
908
909
910
911
912
913
914
915
916
917
918
919
920
921
922
923
924
925
926
927
928
929
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
C
O
R
R
E
C
P
R
O
O
F
[90] W.E. Smith, J. Brownell, C.C. White, Z. Afsharinejad, J. Tsai, X. Hu, S.J. Polyak, X. Gao,
T.J. Kavanagh, D.L. Eaton, In vitro toxicity assessment of amphiphillic polymercoated CdSe/ZnS quantum dots in two human liver cell models, ACS Nano 6
(2012) 9475–9484.
[91] S.Y. Tzeng, L.J. Higgins, M.G. Pomper, J.J. Green, Student award winner in the Ph.D.
category for the 2013 society for biomaterials annual meeting and exposition, April
10–13, 2013, Boston, Massachusetts: biomaterial-mediated cancer-specific DNA
delivery to liver cell cultures using synthetic poly(beta-amino ester)s, J. Biomed.
Mater. Res. A 101 (2013) 1837–1845.
[92] T.R. Arunraj, N. Sanoj Rejinold, N. Ashwin Kumar, R. Jayakumar, Bio-responsive
chitin-poly(L-lactic acid) composite nanogels for liver cancer, Colloids Surf. B
Biointerfaces 113C (2013) 394–402.
[93] K.A. Whitehead, J. Matthews, P.H. Chang, F. Niroui, J.R. Dorkin, M. Severgnini, D.G.
Anderson, In vitro-in vivo translation of lipid nanoparticles for hepatocellular
siRNA delivery, ACS Nano 6 (2012) 6922–6929.
[94] E.E. Hui, S.N. Bhatia, Micromechanical control of cell–cell interactions, Proc. Natl.
Acad. Sci. U. S. A. 104 (2007) 5722–5726.
[95] S. Dragoni, G. Franco, M. Regoli, M. Bracciali, V. Morandi, G. Sgaragli, E. Bertelli, M.
Valoti, Gold nanoparticles uptake and cytotoxicity assessed on rat liver precisioncut slices, Toxicol. Sci. 128 (2012) 186–197.
[96] P.M. van Midwoud, M.T. Merema, E. Verpoorte, G.M. Groothuis, A microfluidic
approach for in vitro assessment of interorgan interactions in drug metabolism
using intestinal and liver slices, Lab Chip 10 (2010) 2778–2786.
[97] R.M. Tostoes, S.B. Leite, M. Serra, J. Jensen, P. Bjorquist, M.J. Carrondo, C. Brito, P.M.
Alves, Human liver cell spheroids in extended perfusion bioreactor culture for
repeated-dose drug testing, Hepatology 55 (2012) 1227–1236.
[98] S.F. Wong, Y. No da, Y.Y. Choi, D.S. Kim, B.G. Chung, S.H. Lee, Concave microwell
based size-controllable hepatosphere as a three-dimensional liver tissue model,
Biomaterials 32 (2011) 8087–8096.
[99] C.G. England, T. Priest, G. Zhang, X. Sun, D.N. Patel, L.R. McNally, V. van Berkel, A.M.
Gobin, H.B. Frieboes, Enhanced penetration into 3D cell culture using two and
three layered gold nanoparticles, Int. J. Nanomedicine 8 (2013) 3603–3617.
[100] V.N. Goral, Y.C. Hsieh, O.N. Petzold, J.S. Clark, P.K. Yuen, R.A. Faris, Perfusion-based
microfluidic device for three-dimensional dynamic primary human hepatocyte cell
culture in the absence of biological or synthetic matrices or coagulants, Lab Chip 10
(2010) 3380–3386.
[101] J.H. Sung, M.L. Shuler, A micro cell culture analog (μCCA) with 3-D hydrogel culture
of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer
drugs, Lab Chip 9 (2009) 1385–1394.
[102] S. March, S. Ng, S. Velmurugan, A. Galstian, J. Shan, D.J. Logan, A.E. Carpenter, D.
Thomas, B.K. Sim, M.M. Mota, S.L. Hoffman, S.N. Bhatia, A microscale human liver
platform that supports the hepatic stages of Plasmodium falciparum and vivax,
Cell Host Microbe 14 (2013) 104–115.
[103] A. Ploss, S.R. Khetani, C.T. Jones, A.J. Syder, K. Trehan, V.A. Gaysinskaya, K. Mu, K.
Ritola, C.M. Rice, S.N. Bhatia, Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures, Proc. Natl. Acad. Sci. U. S. A. 107 (2010)
3141–3145.
[104] N.R. Labiris, M.B. Dolovich, Pulmonary drug delivery, part I: physiological factors affecting therapeutic effectiveness of aerosolized medications, Br. J. Clin. Pharmacol.
56 (2003) 588–599.
[105] P. Zarogoulidis, N.K. Karamanos, K. Porpodis, K. Domvri, H. Huang, W. HohenforstSchmidt, E.P. Goldberg, K. Zarogoulidis, p Zarogouldis, et al., Vectors for inhaled
gene therapy in lung cancer. Application for nano oncology and safety of bio
nanotechnology, Int. J. Mol. Sci. 13 (2012) 10828–10862 (Int J Mol Sci, 13 (2012)
17290-17291).
[106] R.J. Vandebriel, W.H. De Jong, A review of mammalian toxicity of ZnO nanoparticles, Nanotechnol. Sci. Appl. 5 (2012) 61–71.
[107] R. Govender, A. Phulukdaree, R.M. Gengan, K. Anand, A.A. Chuturgoon, Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line, J. Nanobiotechnol. 11 (2013) 5.
[108] Y. Wu, E.K. Shih, A. Ramanathan, S. Vasudevan, T. Weil, Nano-sized albumincopolymer micelles for efficient doxorubicin delivery, Biointerphases 7 (2012) 5.
[109] D. Ding, X. Tang, X. Cao, J. Wu, A. Yuan, Q. Qiao, J. Pan, Y. Hu, Novel self-assembly
endows human serum albumin nanoparticles with an enhanced antitumor efficacy, AAPS PharmSciTech (2013).
[110] N.J. Douville, P. Zamankhan, Y.C. Tung, R. Li, B.L. Vaughan, C.F. Tai, J. White, P.J.
Christensen, J.B. Grotberg, S. Takayama, Combination of fluid and solid mechanical
stresses contribute to cell death and detachment in a microfluidic alveolar model,
Lab Chip 11 (2011) 609–619.
[111] C. Zhang, Z. Zhao, N.A. Abdul Rahim, D. van Noort, H. Yu, Towards a human-on-chip:
culturing multiple cell types on a chip with compartmentalized microenvironments, Lab Chip 9 (2009) 3185–3192.
[112] S. Kumar Mahto, J. Tenenbaum-Katan, J. Sznitman, Respiratory physiology on a
chip, Scientifica (Cairo) 2012 (2012) 364054.
[113] B. Naeye, H. Deschout, V. Caveliers, B. Descamps, K. Braeckmans, C. Vanhove, J.
Demeester, T. Lahoutte, S.C. De Smedt, K. Raemdonck, In vivo disassembly of IV administered siRNA matrix nanoparticles at the renal filtration barrier, Biomaterials
34 (2013) 2350–2358.
[114] A.C. Anselmo, V. Gupta, B.J. Zern, D. Pan, M. Zakrewsky, V. Muzykantov, S.
Mitragotri, Delivering nanoparticles to lungs while avoiding liver and spleen
through adsorption on red blood cells, ACS Nano 7 (2013) 11129–11137.
[115] A. des Rieux, V. Fievez, I. Theate, J. Mast, V. Preat, Y.J. Schneider, An improved
in vitro model of human intestinal follicle-associated epithelium to study nanoparticle transport by M cells, Eur. J. Pharm. Sci. 30 (2007) 380–391.
[116] E.J. Johns, The autonomic nervous system and pressure-natriuresis in cardiovascular–renal interactions in response to salt, Clin. Auton. Res. 12 (2002) 256–263.
D
T
[62] D. Kim, S. Finkenstaedt-Quinn, K.R. Hurley, J.T. Buchman, C.L. Haynes, On-chip
evaluation of platelet adhesion and aggregation upon exposure to mesoporous
silica nanoparticles, Analyst (2014).
[63] N. Korin, M. Kanapathipillai, B.D. Matthews, M. Crescente, A. Brill, T. Mammoto, K.
Ghosh, S. Jurek, S.A. Bencherif, D. Bhatta, A.U. Coskun, C.L. Feldman, D.D. Wagner, D.
E. Ingber, Shear-activated nanotherapeutics for drug targeting to obstructed blood
vessels, Science 337 (2012) 738–742.
[64] E.W. Young, M.W. Watson, S. Srigunapalan, A.R. Wheeler, C.A. Simmons, Technique
for real-time measurements of endothelial permeability in a microfluidic membrane chip using laser-induced fluorescence detection, Anal. Chem. 82 (2010)
808–816.
[65] G. Lamberti, Y. Tang, B. Prabhakarpandian, Y. Wang, K. Pant, M.F. Kiani, B. Wang,
Adhesive interaction of functionalized particles and endothelium in idealized
microvascular networks, Microvasc. Res. 89 (2013) 107–114.
[66] N. Doshi, B. Prabhakarpandian, A. Rea-Ramsey, K. Pant, S. Sundaram, S. Mitragotri,
Flow and adhesion of drug carriers in blood vessels depend on their shape: a study
using model synthetic microvascular networks, J. Control. Release 146 (2010)
196–200.
[67] J.M. Rosano, N. Tousi, R.C. Scott, B. Krynska, V. Rizzo, B. Prabhakarpandian, K. Pant,
S. Sundaram, M.F. Kiani, A physiologically realistic in vitro model of microvascular
networks, Biomed. Microdevices 11 (2009) 1051–1057.
[68] K. Namdee, A.J. Thompson, P. Charoenphol, O. Eniola-Adefeso, Margination propensity of vascular-targeted spheres from blood flow in a microfluidic model of
human microvessels, Langmuir 29 (2013) 2530–2535.
[69] S.P. Samuel, N. Jain, F. O'Dowd, T. Paul, D. Kashanin, V.A. Gerard, Y.K. Gun'ko, A.
Prina-Mello, Y. Volkov, Multifactorial determinants that govern nanoparticle
uptake by human endothelial cells under flow, Int. J. Nanomedicine 7 (2012)
2943–2956.
[70] P. Kolhar, A.C. Anselmo, V. Gupta, K. Pant, B. Prabhakarpandian, E. Ruoslahti, S.
Mitragotri, Using shape effects to target antibody-coated nanoparticles to lung
and brain endothelium, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 10753–10758.
[71] F. Wang, H. Wang, J. Wang, H.-Y. Wang, P.L. Rummel, S.V. Garimella, C. Lu,
Microfluidic delivery of small molecules into mammalian cells based on hydrodynamic focusing, Biotechnol. Bioeng. 100 (2008) 150–158.
[72] G.U. Ruiz-Esparza, J.H. Flores-Arredondo, V. Segura-Ibarra, G. Torre-Amione, M.
Ferrari, E. Blanco, R.E. Serda, The physiology of cardiovascular disease and innovative liposomal platforms for therapy, Int. J. Nanomedicine 8 (2013) 629–640.
[73] S.P. Nordt, A. Minns, C. Tomaszewski, F.L. Cantrell, R.F. Clark, Retrospective review
of digoxin exposures to a poison control system following recall of Digitek(R) tablets, Am. J. Cardiovasc. Drugs 10 (2010) 261–263.
[74] A. Natarajan, M. Stancescu, V. Dhir, C. Armstrong, F. Sommerhage, J.J. Hickman, P.
Molnar, Patterned cardiomyocytes on microelectrode arrays as a functional,
high information content drug screening platform, Biomaterials 32 (2011)
4267–4274.
[75] A. Grosberg, P.W. Alford, M.L. McCain, K.K. Parker, Ensembles of engineered cardiac
tissues for physiological and pharmacological study: heart on a chip, Lab Chip 11
(2011) 4165–4173.
[76] S.R. Shin, S.M. Jung, M. Zalabany, K. Kim, P. Zorlutuna, S.B. Kim, M. Nikkhah, M.
Khabiry, M. Azize, J. Kong, K.T. Wan, T. Palacios, M.R. Dokmeci, H. Bae, X.S. Tang,
A. Khademhosseini, Carbon-nanotube-embedded hydrogel sheets for engineering
cardiac constructs and bioactuators, ACS Nano 7 (2013) 2369–2380.
[77] F. Zanella, R.C. Lyon, F. Sheikh, Modeling heart disease in a dish: from somatic cells
to disease-relevant cardiomyocytes, Trends Cardiovasc. Med. 24 (2014) 32–44.
[78] E. Cimetta, A. Godier-Furnemont, G. Vunjak-Novakovic, Bioengineering heart tissue
for in vitro testing, Curr. Opin. Biotechnol. 24 (2013) 926–932.
[79] H.C. Ott, T.S. Matthiesen, S.K. Goh, L.D. Black, S.M. Kren, T.I. Netoff, D.A. Taylor,
Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial
heart, Nat. Med. 14 (2008) 213–221.
[80] K.R. Stevens, L. Pabon, V. Muskheli, C.E. Murry, Scaffold-free human cardiac tissue
patch created from embryonic stem cells, Tissue Eng. Part A 15 (2009) 1211–1222.
[81] T. Shimizu, M. Yamato, Y. Isoi, T. Akutsu, T. Setomaru, K. Abe, A. Kikuchi, M. Umezu,
T. Okano, Fabrication of pulsatile cardiac tissue grafts using a novel 3-dimensional
cell sheet manipulation technique and temperature-responsive cell culture
surfaces, Circ. Res. 90 (2002) e40.
[82] A. Bussek, M. Schmidt, J. Bauriedl, U. Ravens, E. Wettwer, H. Lohmann, Cardiac tissue slices with prolonged survival for in vitro drug safety screening, J. Pharmacol.
Toxicol. Methods 66 (2012) 145–151.
[83] A. Agarwal, J.A. Goss, A. Cho, M.L. McCain, K.K. Parker, Microfluidic heart on a
chip for higher throughput pharmacological studies, Lab Chip 13 (2013)
3599–3608.
[84] S. Martewicz, F. Michielin, E. Serena, A. Zambon, M. Mongillo, N. Elvassore,
Reversible alteration of calcium dynamics in cardiomyocytes during acute hypoxia
transient in a microfluidic platform, Integr. Biol. (Camb.) 4 (2012) 153–164.
[85] N. Kaplowitz, Idiosyncratic drug hepatotoxicity, Nat. Rev. Drug Discov. 4 (2005)
489–499.
[86] S.R. Khetani, S.N. Bhatia, Microscale culture of human liver cells for drug development, Nat. Biotechnol. 26 (2008) 120–126.
[87] S.B. Leite, I. Wilk-Zasadna, J.M. Zaldivar, E. Airola, M.A. Reis-Fernandes, M.
Mennecozzi, C. Guguen-Guillouzo, C. Chesne, C. Guillou, P.M. Alves, S. Coecke,
Three-dimensional HepaRG model as an attractive tool for toxicity testing, Toxicol.
Sci. 130 (2012) 106–116.
[88] D. Yip, C.H. Cho, A multicellular 3D heterospheroid model of liver tumor and
stromal cells in collagen gel for anti-cancer drug testing, Biochem. Biophys. Res.
Commun. 433 (2013) 327–332.
[89] Y.C. Toh, T.C. Lim, D. Tai, G. Xiao, D. van Noort, H. Yu, A microfluidic 3D hepatocyte
chip for drug toxicity testing, Lab Chip 9 (2009) 2026–2035.
U
N
952
953
954
Q7
955
956
957
958
959
960
961
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
1001
1002
1003
1004
1005
1006
1007
1008
1009
1010
1011
1012
1013
1014
1015
1016
1017
1018
1019
1020
1021
1022
1023
1024
1025
1026
1027
1028
1029
1030
1031
1032
1033
1034
1035
1036
1037
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
E
12
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
1038
1039
1040
1041
1042
1043
1044
1045
1046
1047
1048
1049
1050
1051
1052
1053
1054
1055
1056
1057
1058
1059
1060
1061
1062
1063
1064
1065
1066
1067
1068
1069
1070
1071
1072
1073
1074
1075
1076
1077
1078
1079
1080
1081
1082
1083
1084
1085
1086
1087
1088
1089
1090
1091
1092
1093
1094
1095
1096
1097
1098
1099
1100
1101
1102
Q8
1103
1104
1105
1106
1107
1108
1109
1110
1111
1112
1113
1114
1115
1116
1117
1118
1119
1120
1121
1122
1123
N.S. Bhise et al. / Journal of Controlled Release xxx (2014) xxx–xxx
C
R
O
O
F
P
D
U
N
C
O
R
R
1222
[135] C. Kim, J.H. Bang, Y.E. Kim, S.H. Lee, J.Y. Kang, On-chip anticancer drug test of
regular tumor spheroids formed in microwells by a distributive microchannel
network, Lab Chip 12 (2012) 4135–4142.
[136] C.G. Yang, Y.F. Wu, Z.R. Xu, J.H. Wang, A radial microfluidic concentration gradient
generator with high-density channels for cell apoptosis assay, Lab Chip 11 (2011)
3305–3312.
[137] Z. Xu, Y. Gao, Y. Hao, E. Li, Y. Wang, J. Zhang, W. Wang, Z. Gao, Q. Wang, Application
of a microfluidic chip-based 3D co-culture to test drug sensitivity for individualized
treatment of lung cancer, Biomaterials 34 (2013) 4109–4117.
[138] L. Zhao, J.T. Caot, Z.Q. Wu, J.X. Li, J.J. Zhu, Lab-on-a-Chip for anticancer drug screening using quantum dots probe based apoptosis assay, J. Biomed. Nanotechnol. 9
(2013) 348–356.
[139] S. Jin, K. Ye, Targeted drug delivery for breast cancer treatment, Recent Pat.
Anticancer Drug Discov. 8 (2013) 143–153.
[140] G. Batist, G. Ramakrishnan, C.S. Rao, A. Chandrasekharan, J. Gutheil, T. Guthrie, P.
Shah, A. Khojasteh, M.K. Nair, K. Hoelzer, K. Tkaczuk, Y.C. Park, L.W. Lee, Reduced
cardiotoxicity and preserved antitumor efficacy of liposome-encapsulated doxorubicin and cyclophosphamide compared with conventional doxorubicin and cyclophosphamide in a randomized, multicenter trial of metastatic breast cancer, J. Clin.
Oncol. 19 (2001) 1444–1454.
[141] W.J. Gradishar, S. Tjulandin, N. Davidson, H. Shaw, N. Desai, P. Bhar, M. Hawkins, J.
O'Shaughnessy, Phase III trial of nanoparticle albumin-bound Paclitaxel compared
with polyethylated castor oil-based Paclitaxel in women with breast cancer, J. Clin.
Oncol. 23 (2005) 7794–7803.
[142] S.M. Love, S.H. Barsky, Anatomy of the nipple and breast ducts revisited, Cancer
101 (2004) 1947–1957.
[143] W.C. Dooley, Ductal lavage, nipple aspiration, and ductoscopy for breast cancer
diagnosis, Curr. Oncol. Rep. 5 (2003) 63–65.
[144] M.M. Grafton, L. Wang, P.A. Vidi, J. Leary, S.A. Lelievre, Breast on-a-chip: mimicry of
the channeling system of the breast for development of theranostics, Integr. Biol.
(Camb.) 3 (2011) 451–459.
[145] S.R. Wellings, A hypothesis of the origin of human breast cancer from the terminal
ductal lobular unit, Pathol. Res. Pract. 166 (1980) 515–535.
[146] P.A. Vidi, T. Maleki, M. Ochoa, L. Wang, S.M. Clark, J.F. Leary, S.A. Lelievre, Diseaseon-a-chip: mimicry of tumor growth in mammary ducts, Lab Chip 14 (2014)
172–177.
[147] S. Shah, Y. Liu, W. Hu, J. Gao, Modeling particle shape-dependent dynamics in
nanomedicine, J. Nanosci. Nanotechnol. 11 (2011) 919–928.
[148] J. Tan, S. Shah, A. Thomas, H.D. Ou-Yang, Y. Liu, The influence of size, shape and vessel geometry on nanoparticle distribution, Microfluid. Nanofluid. 14 (2013) 77–87.
[149] M.J. Kim, K. Rhee, Computational analysis of nanoparticle adhesion to endothelium:
effects of kinetic rate constants and wall shear rates, Med. Biol. Eng. Comput. 49
(2011) 733–741.
[150] E. Berthier, E.W. Young, D. Beebe, Engineers are from PDMS-land, Biologists are
from Polystyrenia, Lab Chip 12 (2012) 1224–1237.
[151] K. Schimek, M. Busek, S. Brincker, B. Groth, S. Hoffmann, R. Lauster, G. Lindner,
A. Lorenz, U. Menzel, F. Sonntag, H. Walles, U. Marx, R. Horland, Integrating biological vasculature into a multi-organ-chip microsystem, Lab Chip 13 (2013)
3588–3598.
E
T
[117] B.M. Grabias, K. Konstantopoulos, The physical basis of renal fibrosis: effects of
altered hydrodynamic forces on kidney homeostasis, Am. J. Physiol. Renal Physiol.
(2013).
[118] K.J. Jang, A.P. Mehr, G.A. Hamilton, L.A. McPartlin, S. Chung, K.Y. Suh, D.E. Ingber,
Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity
assessment, Integr. Biol. (Camb.) 5 (2013) 1119–1129.
[119] L. Shintu, R. Baudoin, V. Navratil, J.M. Prot, C. Pontoizeau, M. Defernez, B.J. Blaise, C.
Domange, A.R. Pery, P. Toulhoat, C. Legallais, C. Brochot, E. Leclerc, M.E. Dumas,
Metabolomics-on-a-chip and predictive systems toxicology in microfluidic
bioartificial organs, Anal. Chem. 84 (2012) 1840–1848.
[120] S. Kruetzmann, M.M. Rosado, H. Weber, U. Germing, O. Tournilhac, H.H. Peter, R.
Berner, A. Peters, T. Boehm, A. Plebani, I. Quinti, R. Carsetti, Human immunoglobulin
M memory B cells controlling Streptococcus pneumoniae infections are generated in
the spleen, J. Exp. Med. 197 (2003) 939–945.
[121] C.W. Yung, J. Fiering, A.J. Mueller, D.E. Ingber, Micromagnetic–microfluidic blood
cleansing device, Lab Chip 9 (2009) 1171–1177.
[122] C.H. Heldin, K. Rubin, K. Pietras, A. Ostman, High interstitial fluid pressure — an
obstacle in cancer therapy, Nat. Rev. Cancer 4 (2004) 806–813.
[123] F. Danhier, E. Ansorena, J.M. Silva, R. Coco, A. Le Breton, V. Preat, PLGA-based
nanoparticles: an overview of biomedical applications, J. Control. Release 161
(2012) 505–522.
[124] E.W. Young, Cells, tissues, and organs on chips: challenges and opportunities for
the cancer tumor microenvironment, Integr. Biol. (Camb.) 5 (2013) 1096–1109.
[125] X. Feng, W. Du, Q. Luo, B.F. Liu, Microfluidic chip: next-generation platform for
systems biology, Anal. Chim. Acta 650 (2009) 83–97.
[126] N.S. Bhise, R.B. Shmueli, J.C. Sunshine, S.Y. Tzeng, J.J. Green, Drug delivery strategies
for therapeutic angiogenesis and antiangiogenesis, Expert Opin. Drug Deliv. 8
(2011) 485–504.
[127] D. Wlodkowic, J.M. Cooper, Tumors on chips: oncology meets microfluidics, Curr.
Opin. Chem. Biol. 14 (2010) 556–567.
[128] A. Albanese, A.K. Lam, E.A. Sykes, J.V. Rocheleau, W.C. Chan, Tumour-on-achip provides an optical window into nanoparticle tissue transport, Nat.
Commun. 4 (2013) 2718.
[129] M. Pernia Leal, A. Torti, A. Riedinger, R. La Fleur, D. Petti, R. Cingolani, R. Bertacco, T.
Pellegrino, Controlled release of doxorubicin loaded within magnetic thermoresponsive nanocarriers under magnetic and thermal actuation in a microfluidic
channel, ACS Nano 6 (2012) 10535–10545.
[130] B.J. Kirby, M. Jodari, M.S. Loftus, G. Gakhar, E.D. Pratt, C. Chanel-Vos, J.P. Gleghorn, S.
M. Santana, H. Liu, J.P. Smith, V.N. Navarro, S.T. Tagawa, N.H. Bander, D.M. Nanus, P.
Giannakakou, Functional characterization of circulating tumor cells with a
prostate-cancer-specific microfluidic device, PLoS One 7 (2012) e35976.
[131] M.G. Mauk, B.L. Ziober, Z. Chen, J.A. Thompson, H.H. Bau, Lab-on-a-chip technologies for oral-based cancer screening and diagnostics: capabilities, issues, and prospects, Ann. N. Y. Acad. Sci. 1098 (2007) 467–475.
[132] P. Li, Z.S. Stratton, M. Dao, J. Ritz, T.J. Huang, Probing circulating tumor cells in
microfluidics, Lab Chip 13 (2013) 602–609.
[133] N.T. Elliott, F. Yuan, A microfluidic system for investigation of extravascular transport
and cellular uptake of drugs in tumors, Biotechnol. Bioeng. 109 (2012) 1326–1335.
[134] T. Nawy, Receptive cells feel the squeeze, Nat. Methods 10 (2013) 198.
E
1124
1125
1126
Q9
1127
1128
1129
1130
1131
1132
1133
1134
1135
1136
1137
1138
1139
1140
1141
1142
1143
1144
1145
1146
1147
1148
1149
1150
1151
1152
1153
1154
1155
1156
1157
1158
1159
1160
1161
1162
1163
1164
1165
1166
1167
1168
1169
1170
1171
1172
13
Please cite this article as: N.S. Bhise, et al., Organ-on-a-chip platforms for studying drug delivery systems, J. Control. Release (2014), http://
dx.doi.org/10.1016/j.jconrel.2014.05.004
View publication stats
1173
1174
1175
1176
1177
1178
1179
1180
1181
1182
1183
1184
1185
1186
1187
1188
1189
1190
1191
1192
1193
1194
1195
1196
1197
1198
1199
1200
1201
1202
1203
1204
1205
1206
1207
1208
1209
1210
1211
1212
1213
1214
1215
1216
1217
1218
1219
1220
1221