Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Advances in multicellular spheroids

rsif.royalsocietypublishing.org
formation
X. Cui, Y. Hartanto and H. Zhang
School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia

Review HZ, 0000-0003-4178-6401

Three-dimensional multicellular spheroids (MCSs) have a complex


architectural structure, dynamic cell –cell/cell –matrix interactions and
Cite this article: Cui X, Hartanto Y, Zhang H.
bio-mimicking in vivo microenvironment. As a fundamental building block
2017 Advances in multicellular spheroids
for tissue reconstruction, MCSs have emerged as a powerful tool to
formation. J. R. Soc. Interface 14: 20160877. narrow down the gap between the in vitro and in vivo model. In this
http://dx.doi.org/10.1098/rsif.2016.0877 review paper, we discussed the structure and biology of MCSs and detailed
fabricating methods. Among these methods, the approach in microfluidics
with hydrogel support for MCS formation is promising because it allows
essential cell –cell/cell –matrix interactions in a confined space.
Received: 1 November 2016
Accepted: 11 January 2017

1. Introduction
Traditionally, cell culture on the two-dimensional tissue plastics has been used
Subject Category: for cell biology studies and disease models for drug screening with the assump-
Reviews tion that monolayer cells reflect the physiology of real tissues. However, in such
a two-dimensional platform, cells are typically exposed to a rigid solid surface
Subject Areas: on the basal side and to a liquid at the apical surface. Inhabiting such a
two-dimensional rigid substrate requires cytoskeletal adjustments by the
biomedical engineering, bioengineering,
surviving cells, because they lack exposure to the extracellular matrix (ECM)
biotechnology that is unique to each cell type, which may produce counterfeit polarity and
cause abnormal cell metabolism and protein expression [1]. Furthermore,
Keywords: two-dimensional systems cannot provide a complex and dynamic microenvir-
multicellular spheroids, tissue engineering, onment for cells, and thus lead to spurious findings to some extent by
three-dimensional culture, microfluidics, forcing cells to adjust to an artificial and rigid surface. Studies have shown
hydrogel that two-dimensional cell culture cannot replicate real microenvironment and
cell behaviours in vivo because of lack of cell –cell and cell –matrix interactions
and loss of tissue-specific architecture, mechanical and chemical cues, which are
essential for unique functions of real tissues in the human body. For instance, a
Author for correspondence: decrease in b1-integrin hindered multicellular spheroid (MCS) formation of
H. Zhang PC3 prostate adenocarcinoma cells, while the decrease was not found in the
e-mail: hu.zhang@adelaide.edu.au two-dimensional culture [2]. Therefore, a new platform for in vitro cell culture
has been pursued and three-dimensional cell culture has been demonstrated
to recreate the in vivo microenvironment and physiological relevance [3].
A number of three-dimensional culture models have been explored. Three-
dimensional MCS is an excellent in vitro three-dimensional model, mimicking
the in vivo processes, such as embryogenesis, morphogenesis and organogen-
esis [4]. MCSs are cell aggregates that have complex cell-to-cell adhesion and
cell-to-matrix interaction, which result in gradient generation for nutrients,
gases, growth factors and signal factors. This structure recapitulates the
microenvironment of cells that has been observed in the real tissues.
MCSs were first fabricated by Moscona & Moscona through self-assembling
of cell suspensions, and they found these tissue-like aggregates were able to
restore characteristics in the original tissue [5]. Since then, methods have been
developed to form MCSs. Furthermore, a range of cells have been explored to
form MCSs, including cancer cells [6], mesenchymal stem cells and human
pluripotent stem cells [7]. MCSs exhibit similar features in the in vivo physio-
logical conditions [8], for example, cardiomyocyte spheroids beat with heart-
like rhythms [9], hepatocyte spheroids perform liver-like functions [9], and

& 2017 The Author(s) Published by the Royal Society. All rights reserved.
human endothelial cells vascularize fibroblast microtissues growth and differentiation of tumour spheroids, while 2
[10]. MSCs are considered to be the building blocks to bio- stroma contributes to tumour progression. Normal stroma

rsif.royalsocietypublishing.org
fabricate three-dimensional functional living macrotissues delays or prevents MCS formation but the abnormal one pro-
and organ constructs via organ printing, which could be a motes tumourgenesis [24]. Hence, stroma plays a crucial role
paradigm shift for tissue engineering [11]. Therefore, MCSs in the MCS morphology and the evolution of tumour spher-
are considered as a bridge to bring in vitro and in vivo oids. Additional stroma elements promote tumour spread
together, and they are becoming an emerging biomedical and invasion [25] and stroma composition or structure may
tool in many areas. This paper delivers a comprehensive be altered when the function of vascular cells or fibroblasts
review on the methods of MCSs fabrication. Special foci changes, or the influx of inflammatory cells increases [26].
are devoted to recent progress in MCSs formation in the The interaction between endothelial and tumour cells in fab-
last 5 years. ricated MCSs offers information on formation of blood
capillaries in tumours.

J. R. Soc. Interface 14: 20160877


In vitro MCSs have been fabricated with different struc-
2. Formation mechanism and structure of tures. The most common one is a three-layered structure
with cells in asynchronously proliferative, quiescent and
multicellular spheroids necrotic states (figure 3a). The proliferative rim contains
MCS formation may be due to cell adhesion and/or cell cells with intact nuclei and abundant microvilli and these
differentiation. It has been reported that MCS formation cells are active in proliferation and metabolism. Cells with
involves three critical steps [12]. (i) Dispersed cells initially shrunk nuclei and sunken-in membranes are distributed in
are drawn closer to form loose aggregates due to their the quiescent stage and these cells have minimum metabolic
long-chain ECM fibres with multiple RGD motifs that can activities but become active after exposure to nutrients. Cells
bind tightly to the integrin on the cell membrane surface. in the necrotic core have disintegrated nuclei and membranes
Direct cell –cell contact due to initial aggregation results in and they lose their activities because of starvation of nutrients
upregulated cadherin expression. (ii) Cadherin is accumu- and accumulation of toxic wastes. The layer thickness is
lated at the membrane surface. (iii) Cells are compacted into dependent on the size of the spheroids. The diffusion limit-
solid aggregates to form MCSs due to the homophilic cad- ation of most small molecules like oxygen in MCSs is
herin –cadherin binding [13] (figure 1). The ECM fibres and around 150–200 mm, which is similar to that in the avascular
cadherin type and concentration may vary for different tissue and tumour mass in vivo. Beyond the diffusive limit,
type of cells [14–16]. For example, E-cadherin is responsible metabolic wastes are accumulated in the inner layer of
for tight packing of MCF7, BT-474, T-47D and MDA-MB- MCSs [30 –32], leading to a necrotic core. Proton magnetic
361 cells [15]. It was reported that b-catenin sequestrated by resonance with pH-sensitive indicators [32] as well as micro-
E-cadherin can initiate transcriptional activation of proteins electrodes [31,33] have been used to study the transportation
such as cyclin D1 and c-Myc, which control the G1 to S- of nutrients, wastes and oxygen transportation and confirm
phase transition in the cell cycle to help cell proliferation the concentration gradients inside the MCS.
which also result in cell differentiation within MCSs [17]. A vascular lumen-like hollow structure has also been
Meanwhile N-cadherin mediates the spontaneous formation observed for endothelial MCSs due to the apoptosis caused
of spheroids in MDA-MB-435S [15]. MDA-MB-231 and SK- by central cell polarization [34]. A unique phenotype of
BR-3 cells form spheroid structure due to collagen I/integrin MCSs formed when bovine aortic endothelial cells grew
b interaction without cadherin involvement [18]. within a matrix that was full of basal membrane constituents.
Intercellular adhesion proteins such as connexin and pan- A polarized epithelium was established in the outermost
nexins that influence cell– cell contacts contribute to the layer with an apical surface pointing inside and a lumen
formation of MCSs. As a class of gap junction proteins [19], was formed due to gradual loss of cells in the sphere interior.
pannexins were introduced into the C6 glioma cells in The spheroid structure obtained from human mammary
order to accelerate the MCS formation process and obtain a epithelia cells [35,36] have been used for studying apoptosis
more mature F-actin cytoskeleton. The result showed pan- and tissue morphogenesis. This model is also employed for
nexin 1 as the conduit for ATP release initiated a signalling investigating the oncogenic pathway to affect cellular cycles
that dramatically accelerated the assembly of large MCSs [20]. and biochemical pathways to transmit signals from cell –
The cytoskeleton also plays an important role in MCS for- matrix contact in order to increase the cell survival rate.
mation (figure 2, the actin filaments in the cytoskeleton of the Heterotypic cell-to-cell interactions play an important role
spheroids [21]). The actin filaments undergo significant in maintaining the hierarchy architecture and physiological
changes during MCS formation. The expanded microfila- functions of tissues and heterotypic MCSs are generated for
ments as stress fibres become localized along the cell mimicking the architecture and functions of real tissues. The
periphery. The cytoskeleton as a force generation structure heterotypic MCSs have been generated from co-culturing
performs as a continuous pre-stressed lattice that keeps cellu- fibroblasts ( parenchymal hepatocytes with human dermal
lar structural stability. Morphogenetic phenomena promote fibroblasts, rat hepatocytes with NIH/3T3 fibroblasts)
the emergence of ordered structures, resulting in the MCSs [37,38], pancreatic islet cells with hepatocytes [39] or bone
formation [22]. marrow stoma cells with hepatocytes [40]. Different architec-
Tumour MCSs in vivo have two major components: one is tures for normal and malignant breast epithelial cells have
proliferating neoplastic parenchymal cells, and the other one been fabricated to simulate the tumour micro-environment
is supportive stroma, constituting half the mass of most [41]. Co-culture of tumour cells with endothelial cells allows
malignant tumours and including fibroblasts, dendritic building small capillaries within the MCS structure to avoid
cells, lymphocytes, blood vessels, macrophages as well as a necrotic or quiescent core in the centre. Transportation of
other myeloid cells [23]. The parenchymal cells determine nutrients and oxygen from the outer layer to the centre by
3

integrim–ECM cadherin expression cadherin–cadherin

rsif.royalsocietypublishing.org
interactions and accumulation interactions

J. R. Soc. Interface 14: 20160877


cell aggregation spheroid compaction

Figure 1. MCS formation process. Cells establish loose bonds through integrin– ECM. A delay stage is followed for cadherin expression and accumulation on the
extracellular surface. A compact MCS forms due to homophilic cadherin – cadherin interactions. (Online version in colour.)

the endothelial cells is realized, which mimics the transpor-


tation in the in vivo tumours. MCSs were co-cultured with
3. Conventional multicellular spheroid formation
macrophages or lymphokine-activated killer cells in order to from dispersed cells
measure the cytotoxic and cytostatic activity and examine the In vitro production of MCSs has several challenging issues.
immigration of the immune cells [42]. It was found that lactic Economic production of MCSs in a short time frame needs
acid generated from MCSs disturbed the migration of mono- to be considered as the first criteria for choosing an approach
cytes into MCSs, working as a potent immune suppressor for for MCS formation. The ability to produce uniform-size
monocytes/dendritic in the tumour milieu. Tumour cell inva- MCSs is also crucial. Damage of MCSs and the impact on
sion and metastasis have also been investigated through the physiology should also be addressed. Many approaches
interactions between tumour cells and fibroblasts [43,44]. Fur- have been developed to fabricate MCSs, such as dispersed
thermore, epithelia cells in a heterotypic spheroid have been cells (referred to as ‘matrix-free’ herein), cells embedded in
used to investigate cell –cell interaction for different types of hydrogel matrix (referred to as ‘matrix-based’ herein) and
cells [45]. A spheroid formed by co-culturing mesenchymal microfluidic platforms as shown in figure 4. The matrix-free
stem cells and primary liver cells behaved like hepatocyte approach has been commonly used to produce MCSs,
cells, and was explored for the heterotypic cell co-culture for such as suspension culture in non-adhesive surfaces
cell proliferation and differentiation with the potential for [18,50 –52], hanging drops [53,54], spinner flasks or rotational
regenerative medicine since the co-culture system was more bioreactors [55–57] and external-force-driven MCS aggrega-
efficient in inducing differentiation of mesenchymal stem tion [21,58 –62]. These methods have been detailed and
cells into hepatocyte-like cells [46]. reviewed in previous papers [24,63,64]. A summary of the
The microenvironment of MCSs is crucial for cell beha- matrix-free approach can be seen in table 1. The approach
viours and functions. The MSCs model has been used to of using dispersed cells in microfabricated chambers for
investigate the impact of the architectural, physical and phys- forming MCSs is discussed in more detail in §5.
iological microenvironment on the tumorigenic phenotypes. Overall, conventional approaches are easy, quick and cheap
Intracellular gene expression is influenced by altering the to operate. However, some approaches, such as hang drops, are
chemical composition of the extracellular microenvironment labour consuming. In addition, it is quite challenging to control
[24]. Abnormal cell behaviours such as tumour progression the size of the spheroid and to avoid shear damage to MCSs.
are caused by inappropriate cell –microenvironment inter- Lack of matrix support results in no cell–matrix interactions
actions [47]. Martin et al. [48] reported that cells performed that is essential in MCSs cell biology and functions.
normal embryogenesis if they were located in the uterus,
while they were differentiated into malignant tissue cells
when they were co-cultured with ectopic embryonic cells.
Oral squamous carcinoma cells had similar behaviours in
4. Matrix supported multicellular spheroid
the synthetic poly (lactide-co-glycolic acid) (PLGA) scaffold formation
in vitro and in vivo compared to two-dimensional culture as Hydrogel networks as scaffolds provide bio-mimicking
evidenced by increase in secretion of vascular endothelial matricies to MCSs to induce cell –matrix interactions for fab-
growth factor (VEGF), interleukin 8 (IL-8), as well as basic rication of MCSs. The scaffold structure, morphology and its
fibroblast growth factor (FGF). An overall increase in the components can be adjusted for tuning the microenviron-
VEGF and FGF expression was able to mimic that in the ment for MCSs so that the cellular responses to the
in vivo tumours. The results also highlight that some factors microenvironment can be monitored during MCS formation
were found in the three-dimensional culture but not in the [65] (table 2).
two-dimensional culture, such as IL-8. Through analysis of
the cumulative percentage of these factors, IL-8, instead of
VEGF, was proven to play the most important role in angio- 4.1. Natural polymers
genesis, which has changed our understanding of the role of Natural scaffolds from hydrogels, such as collagen, Matrigel
IL-8 from the two-dimensional study [49]. and chitosan (CS), have been used to culture MCSs. Other
4
(a) (b) (c) (d)

rsif.royalsocietypublishing.org
J. R. Soc. Interface 14: 20160877
Figure 2. The confocal images of the BT-474 spheroid [21]: (a) nuclei, (b) focal contacts (antivinculin antibody), (c) F-actin and (d ) merged. The scale bar is
100 mm. (Online version in colour.)

natural polymers, such as agar for ovarian cancer cellular HA scaffold than in two-dimensional culture, which was
MCSs [1], fibrin [82] and silk fibroin protein [87] for B16-F1 similar to the LNCaP growth in vivo. Human glioblastoma
cell and breast cancer cell line MDA-MB-231 cells, respectively, U373-MG and U87-MG cells were found to form heterotypic
have also been explored. Natural hydrogels are favourable MCSs after culturing them inside the HA scaffold for 24 h
for cell proliferation due to its enriched ECM proteins [70]. MDA-MB-231 and MDA-MB-468 cells mixed with the
or mimicking ECM components. They are also highly HA hydrogel were injected into mice to mimic the in vivo
biocompatible [88]. environment for tumour spheroid formation, and HA was
Collagen is a main structural protein in human tissues. found to stimulate cell proliferation and reduce the variabil-
Collagen is often used as an excellent matrix for cell ity in the tumour size. This hydrogel was also found to
attachment due to its integrin binding sites [89]. The three- facilitate tumour–tissue integration. Necrosis inside the
dimensional porous structure of collagen hydrogels allows formed MCSs was reduced, and vascularization in the
sufficient exchange of oxygen, wastes and nutrients inside structure was improved [71].
the scaffold [90] for short-term or long-term MCS culture CS is the second most abundant material that can be pro-
[91]. The biodegradability due to enzymatic reactions offers duced from crustacean, molluscs, squid and insects. The
a simple method of harvesting MCSs from the scaffold. biodegradability by lysozyme on the b-1, 4 glyosidic linkages
Human embryonic stem cells (hESCs) were introduced into enables mature MCS to release from the CS-based scaffold
the collagen scaffold, and after 5 days of culture hESCs [93]. A porous CS scaffold was attempted to culture MCF-7
were differentiated into hepatocytes. The hepatocytes cells for producing MCSs and the scaffold with a high
formed MCSs which possessed all characteristics and func- degree of deacetylation was found to improve cancer cell
tions of human hepatocytes in vivo [66]. Luminal cells were attachment and proliferation [72]. However, very low solubi-
co-cultured with myoepithelial cells and fibroblasts in the col- lity of CS in water due to its strong hydrogen bonding
lagen type I matrix, and after 7 days of culture heterotypic hampers its wide application in MCS formation. CS co-
MCSs were formed inside the matrix [67]. Fang et al. [68] polymers have been synthesized to improve the solubility.
cross-linked collagen with Matrigel to create a hydrogel scaf- CS-alginate was synthesized as a hydrogel scaffold for
fold to culture human breast carcinoma cells (MDA-MA-231) culturing prostate cancer cell MCSs [94]. This co-polymer
and colorectal carcinoma cells (HCTT116) for forming MCSs. was more biocompatible, biodegradable and less immuno-
The new hydrogel had better biocompatibility and repro- genic [95]. In addition, its chemical structure is similar to
duced the in vivo solid tumour microenvironment. One glycosaminoglycans (GAGs), a vital part of MCS ECMs.
unique method of using collagen microparticles with a size Instead of a hydrogel scaffold, CS-alginate was also fabri-
of 10 –20 mm to fabricate functional spheroids was reported cated as a fibrous scaffold coated with collagen to culture
[92]. The collagen micro-particles were fabricated via droplets MCF-7 cells. Its porous structure optimally replicated the in
in a non-equilibrium state in a micro-channel or membrane vivo environment [96]. Small aggregates were observed after
emulsification. Agarose-coated microchambers were used 2 days of culture. The size of spheroids was up to more
to prepare heterogeneous composite spheroids composed of than 100 mm after 6 days. Cell growth within this scaffold
hepatocytes and collagen particles. This approach recreates presented a spatial growth pattern with an improved
a similar in vivo microenvironment due to the incorporating growth rate and drug resistance. Apart from co-polymerized
ECM component collagen in the MCSs. with natural polymers, CS is also combined with synthesized
Hyaluronic acid (HA)-based hydrogels are also applied polymers for MCS formation. For instance, a poly(L-glutamic
in MCS culture. Natural HA is found in the ECM of malig- acid) and CS scaffold was developed to culture MCSs from
nant tumours, and it promotes cancer cell proliferation and adipose-derived stem cells (ADSCs) for cartilage regeneration
spheroid formation [69] because the excellent viscoelasticity shown in figure 5. This scaffold was repulsive to protein
of the HA hydrogel is able to mimic the stiffness of the natu- adsorption and facilitated ADSC MCSs formation by mini-
ral environment. Prostate cancer cells (LNCaP PCa) were mizing cell –matrix attachment to promote cell –cell
seeded into the HA hydrogel system, and after 7 days of cul- contacts. MCSs of 100 mm were observed after 2 days of cul-
ture MCSs were harvested with a size of 100 mm, and ture [73]. ASCs spheroids grown in this scaffold improved
showed a significant upregulation of VEGF165 and IL-8 chondrogenic differentiation and dramatically decreased the
expression. LNCap had a lower proliferation rate in the deposition of collagen type I.
(a) (b) (c) 5

rsif.royalsocietypublishing.org
necrotic nutrients (O2)
zone ATP levels

+ +

waste (CO2)
100 mm

J. R. Soc. Interface 14: 20160877


proliferating lactase levels
zone
quiescent zone

Figure 3. Microenvironment and structure of an MCS. (a) MCS has a spherical geometry with different regions for proliferating, quiescent and dead cells. Different
mass transport rates for nutrient, O2, ATP, waste, CO2 and lactase are presented in the multilayer structure. (b) The live/dead image indicates the morphology of an
MCS, green represents live/proliferating cells ( proliferating zone), while red represents dead cells (quiescent and necrotic zone) [27]. (c) The scanning electron image
of a human epithelial carcinoma A431 MCS grown for 3 days [28]. The smooth profile shows the final stage of spheroids formation. Cadherin – cadherin interactions
lead to the deposition of ECM. As a result, a compact, smooth profile of the spheroid can be observed without distinguishable cell morphology [29]. (Online version
in colour.)

Matrigel is derived from the basement membrane of Poly(ethylene glycol) (PEG) is one of the most popular
Engelbreth-Holm-Swarm(EHS) mouse tumour cells. Some polymers for three-dimensional cell culture due to its non-
key components for cell growth and attachment such as col- toxicity and non-immunogenicity. PEG is cross-linked via
lagen and laminin are found in Matrigel [97]. As one of the various means such as photo-polymerization and emulsion
popular ECM protein-enriched hydrogels, Matrigel has polymerization. PEG was cross-linked with other polymers
been widely used for MCS formation from PC-3M, PrCa such as poly(ethylene oxide) (PEO) to enhance polymer net-
and NCI-H600 cells [74]. The comparison of HepG2 MCS cul- work performance for MCS fabrication. Through adjusting
ture in collagen, gelatin and Matrigel hydrogels revealed the PEG amount in the co-polymer, PEG-based hydrogels
cells proliferated rapidly in Matrigel, and larger MCSs were with different mechanical properties were used to culture
harvested from Matrigel [75]. Moreover, a Matrigel with hepatocellular carcinoma cell line (Huh7.5) for formation of
lung cancer cell mixture injected into an animal model MCSs to explore the impact of microenvironmental stiffness
resulted in rapid formation of tumour MCSs in animals com- on cell aggregations [12]. Larger spheroids formed within
pared with cell injections without Matrigel. The similar the hydrogel network with better compliance or lower stiff-
results were also obtained from cancer cells of A253 and ness. Furthermore, cell proliferation, albumin secretion and
B16F10 [76]. However, three-dimensional tumour models CY450 expression in spheroids were found to perform
grown on the Matrigel exhibited less similarity to the better within compliant hydrogels, which may be due to
in vivo tumours compared with three-dimensional poly (lac- better diffusion of oxygen and nutrients within a more
tide-co-glycolide) (PLG) engineered tumours because of a compliant matrix. PEG-based hydrogels are formed via two
decreased level of IL-8 expression in the Matrigel [49]. There- different polymerization approaches: chain addition metha-
fore, it is important to choose appropriate three-dimensional crylate-based and step-growth thiolene polymerization,
physical supports for fabricating MCSs. were used for submandibular glands (SMG) MCS growth
The natural polymers share the structural or composition and formation. Step-growth thiolene polymerization had
similarity with normal tissue ECM, have rich components, better performance for cell viability, proliferation and cell
meet the amplified extracellular signalling needs and support aggregation due to the reduced membrane peroxidation
cell behaviours. The impediments of using natural polymers and intracellular reactive oxygen species formation [83].
for MCSs are the accessibility of the materials for the tissues Because of its excellent biodegradability, PLG has been
of interest, dreary methodology and undesirable remnant widely investigated for three-dimensional cell culture. The
proteins and confounding signalling proceedings [98]. In key component of this synthesized polymer is extracted
the case of multiple-component scaffolds, additional compo- from natural metabolites and it has great biocompatibility.
sition may be required for optimal support of cell growth. PLG has been used in different forms such as foam, fibres
and sponges [100]. A porous PLG microsphere scaffold was
developed to culture hepatocyte spheroids [84]. This
4.2. Synthesized polymers approach accelerated MCS formation and the porous scaffold
Synthesized polymers have better structural complexity and structure maximized cell attachment and transportation of
design flexibility that are tuned to mimic the in vivo nutrients, oxygen and wastes. MCF-7 and U87 cell lines
environment for MCS culture and formation. In contrast were cultured in the PLG scaffold to form MCSs, and the
to natural polymers, synthesized polymers are able to be PLG scaffold exhibited good performance in recreating the
modified to have desired ECM characteristics for the cell microenvironment for tumour engineering [43]. The tumour
type of interest to promote cell aggregation and maintain model obtained from the PLG scaffold possesses similar
tissue functionality [99]. microenvironmental characteristics to in vivo tumours. The
(a) (b) (c) (d) 6

rsif.royalsocietypublishing.org
(e) (f) (g)
V
S

J. R. Soc. Interface 14: 20160877


Figure 4. Methods for MCS formation. (a) Forced floating, (b) hanging drop, (c) spinning flask, (d ) rotating vessel, (e) electrical-force assisted, ( f ) magnetic-force
assisted and (g) matrix-based.

expression of IL-8, an angiogenetic factor, was found to be Ewing’s sarcoma cells to form MCSs and the MCSs exhibited
upregulated in cells cultured in the PLG scaffold [99]. The greater chemo-resistance and different gene expressions from
angiogenic feature was mapped with that in the in vivo two-dimensional culture [86]. MCSs from breast, prostate and
tumours, and cells in this model were less sensitive to che- Lewis lung cancer cells were successfully demonstrated in the
motherapy, which demonstrated the yielded tumours had poly(lactic-co-glycolic acid)(PLGA) scaffold [99].
improved malignant potential. PLG was mixed with hydro- Overall, synthesized polymers show great design flexi-
xyapatite (HA) to replicate a bone-like environment to bility through co-polymerizing with other functional
culture breast cancer spheroids [101]. This scaffold promoted monomers. The physical and chemical properties are tuned
breast cancer cell proliferation and aggregation because HA to mimic the microenvironment for MCS formation. In
encouraged the neoplastic and metastatic growth of breast addition, different from natural polymers with variations
carcinoma cells and stimulated IL-8 secretions. from batch to batch, synthesized polymers have high repro-
Poly(N-isopropylacrylamide) (PNIPAM) hydrogel is ducibility and improved handling characters. However,
another popular polymer network due to its thermal-reversi- their toxicity and degradability are two major concerns in
bility which allows harvesting of MCSs without toxic or the application of MCS formation.
potent chemicals. Through co-polymerization with other One of the challenging issues is to release MCSs from
monomers, the resultant copolymers can accommodate differ- natural or synthesized polymers. Most natural polymers
ent types of cells, promote cell proliferation and aggregation, except alginate may be degraded by introduction of specific
and maintain tissue functionality. This PNIPAM network enzymes. However, it may be necessary to remove any
was used for culture of HepG2 MCSs and a microgel of enzyme residues for certain biomedical applications. The
around 300 nm was found to have better cell proliferation alginate matrix becomes weak in an acidic environment due
and MCS formation [102]. The PNIPAM polymer was modi- to gradual loss of ironic bonding and eventually the matrix
fied by acrylic acid (AA) for culturing HepG2 MCSs. is degraded to release MCSs. Synthesized polymers normally
PNIPAM-AA exhibited less shrinkage for long-term culture request toxic chemicals to break chemical bonds within their
and maximally maintained the scaffold structure, and polymer structures. Thermal-sensitive polymer matrices can
HepG2 cells proliferated best in the hydrogel with 1% AA in easily release spheroids by manipulating the cell culture
the copolymer [85]. PNIPAM-AA microgels were further galac- temperature, which minimizes potential damage to MCSs.
tosylated to culture HepG2 MCSs. The galactose ligands As a result, thermally sensitive polymers may be considered
helped HepG2 MCSs in performing liver-specific functions one of best candidates as a matrix to recover spheroids.
[6]. PEG was also introduced to PNIPAM hydrogel for better
cell attachment. Human pluripotent stem cells (hPSCs) were
cultured in the hydrogels to form spheroids. hPSC-derived 5. Multicellular spheroids formation on the
spheroids showed high proliferation, maintained the pluripo-
tency in suboptimal culture conditions and had a high survival microfluidic platform
rate [7]. pNIPAM-PEG hydrogel was also used to culture The microenvironment that comprises complex chemical and
HepG2 spheroids; using this method, HepG2 cells were pre- mechanical cues is one of the most important factors for form-
aggregated before they were cultured in the hydrogel [103]. ing MCSs. Specific physico-chemical properties, such as
HepG2 cells were treated by NaIO4 to produce surface alde- oxygen tension, temperature, pH, osmolality and local con-
hyde functionalities, and they were aggregated in the centration of soluble factors, can significantly influence cell-
presence of an inter-cell linker, acrylic acid-modified CS. This to-cell and cell –matrix interactions [104]. Conventional
method is very rapid, taking 1 day to form compact spheroids. approaches for forming MCSs are less flexible in tuning the
Others polymers have also been reported for MCS for- microenvironment and performing temporal and spatial
mation. Poy(1-carpolacton) (PCL) was used to culture TC-71 stimuli on cells. Microfluidics, as an emerging tool to control
Table 1. Matrix-free approach for MSCs fabrication. 7

rsif.royalsocietypublishing.org
methods advantages disadvantages cell type reference

forced- poly(HEMA) —inexpensive cost —variations in MCS size MCF7-ADR,T47D, MCF-7 [18]
floating on coating —simple procedure and shape
non- agarose coating —easy accessibility —intensive labour HeLa [50,51]
adhesive poly (N-p- —reproducibility requirement 3T3colne A31, Type II lung [52]
surfaces vinlbenzyl-D- —large-scale production —lack of cell– matrix alveolar cells
lactonamide) interactions

J. R. Soc. Interface 14: 20160877


coating
hanging drop multi-wells —uniform-size MCSs —intensive labour HepG2, MCF-7 [53]
super —easy operation requirement L929, SaOs-2 [54]
hydrophobic —high reproducibility —small MCSs
chip —controlled environment —incompetency for mass
production
bioreactor spinner flask —simple culture method —variations in MCS size Chinese hamster V79 [55,56]
rotational culture —mass production and shape PC3, LNCaP, DU 145, BT20 [57]
(NASA) —long-term culture —shear damage of MCSs
—dynamic controlled —lack of cell– matrix
environment interactions
—excellent mixing for
nutrient and oxygen
delivery, and waste
removal
force driven magnetic force —controllable MCS sizes —loss of aggregate NIH/3T3, human umbilical [58,59]
and shapes integrity after removal of vein endothelial cells,
—applicability to a variety external forces bone marrow-derived
of cell types for forming —damage of cell human mesenchymal
MCSs morphologies due to stem cells
electric force —co-culture of two or extra forces Jurkat cells [60,61]
acoustic force more different types of —lack of cell– matrix HepG2, BT474 [21,62]
cells for MCS formation interactions
—rapid formation

the flow within micrometre-scale channels, is capable of reported to be 100–150 mm because of lack of oxygen [11]. A
manipulating the parameters dynamically and spatially, gas-permeable spheroid culture chip was developed to con-
thus creating unique environments to meet the requirements tinuously supply oxygen to spheroids during culture [106].
for MCS formation. In addition, microfluidics can reduce As a result, the maximum size of spheroids was able to
the shear stress in order to minimize cell damage, and reach 600 mm with enhanced cell proliferation, viability, meta-
micrometre-scale chambers inside the microfluidics can be bolic capacity as well as albumin secretion rates.
manipulated to control the size of MCSs [105]. Furthermore,
integration of analytical tools with microfluidics (figure 6), 5.1. Micro-moulding
such as Raman, fluorescence spectrometry and UV-Vis spec- The micro-moulding method has been explored to generate
trometry, enables a rapid, in situ and dynamical analysis MSCs by fabricating complex structures using the lithogra-
during MCS formation. To screen optimized culture con- phy technique [109,110]. It has been documented that this
ditions for forming MCSs from rare cells or primary cells the micro moulding method can significantly reduce consump-
microfluidics, consumption of chemical or biochemical tion of reagents, create desired concentration gradients of
reagents is reduced since microfluidics often requires a very growth/signal factors and nutrients, allow high-density
small amount of liquid [108]. Furthermore, oxygen supply to culture at a high cell-to-fluid volume ratio and decrease
MCSs can be significantly improved through microfluidic shear stress under the laminar condition in micro-scale
chips. In conventional approaches, oxygen depletion in the structures [111].
centre core of a large spheroid leads to hypoxic conditions Du et al. [112] proposed a method to fabricate different
that cause cell necrosis at the heart of spheroids. Therefore, micro-scale structures with PEG-based cell laden hydrogels
the maximum size of spheroids consisting of viable cells was to form MCSs from NIH 3T3 mouse fibroblasts. The
Table 2. Typical polymers used as matrix supports for MCSs fabrication. 8

rsif.royalsocietypublishing.org
polymer type features for MSC formation cell types

natural collagen —protein-based hydrogels for better cell hESCs, hepatocytes [66], luminal cells [67],
polymers attachment MDA-MA-231, HCTT116 [68], HepG2 [60]
—weak mechanical strength
HA- —excellent viscoelasticity LNCap [69], U373-MG, U87-MG [70], MDA-MB231,
—promoting cell proliferation MDA-MB-468 [71]
CS —carbohydrate structure similar to the ECM MCF-7 [72], adipose-derived stem cells [73]

J. R. Soc. Interface 14: 20160877


component GAGs for better cell attachment
—insoluble in aqueous solution at
physiological conditions
Matrigel —rich in growth factors, signal factors for cell PC-3M, PrCa, NCI-H600 [74], HepG2 [75], A235,
proliferation B16F10 [76]
—expensive and animal derived
agar —damage on cells by forces from gel human foreskin dermal fibroblasts [77], cortical cell
formation inside the agar [78]
—excellent mechanical properties
alginate —rapid formation of hydrogel scaffolds human fat-derived stromal vascular fraction cells
—strong chemicals required to break hydrogel [79], human colon cancer cells (HCT116) [80]
networks to harvest MCSs
fibrin —promoting cell attachment human mesenchymal stem cell [81], B16-F1
—high cell viability cell [82]
synthesized PEG —good biocompatibility Huh7.5 cells [12], submadibular glands [83]
polymers —irreversible gelation leading to challenges in
releasing MCSs from gel
PLG/PLGA —biodegradable and biocompatible hepatocytes [84], MCF-7, U87 [49]
NIPAM-based —thermally reversible to release MSCs during HepG2 [85], HeLa [51], human pluripotent stem
hydrogel harvest cells [7]
—low viscoelasticity
—porous structure with great nutrient,
oxygen and waste transport
poly(1- —high porosity TC-71 Ewing’s sarcoma cells [86]
carpolacton) —a large surface area-to-volume ratio for
cellular attachment
—high reproducibility

hydrophobic effects drove the ‘lock and key’ assembly of collagen/Matrigel mixture containing HepG2 were loaded
microgels to form cross- or rod-shaped structures. Three- into a mould from the PDMS membrane. After gelation, gel
dimensional tissue constructs containing MCSs were gener- blocks were transferred into the culture medium for further
ated from these structures. Micro-patterned methacrylated forming MCSs. The MCS size was controlled by the size of
HA hydrogels were applied to fabricate similar MCSs from the gel blocks.
NIH 3T3 mouse fibroblasts [29]. A patterned polydimethylsi- A ‘bottom-up’ fabrication approach was developed for
loxane (PDMS) stamp was used to hold the HA precursor macroscale three-dimensional structures [115]. Collagen
solution mixed with cells and the photo-initiator (figure 6a). beads containing HepG2 cells were generated from an axi-
After exposure to UV light, the HA solution was solidified symmetric flow focusing device in a PDMS mould. The
and the PDMS mould was removed. The size and shape of cell-laden beads were stacked to form a complex millimetre-
MCSs were manipulated by the geometry of the PDMS thick tissue. After a couple of hours, the beads made contact
mould. In addition, HA was biodegradable by enzyme and with each other and were compacted into a designed shape.
MCSs were released from the block during the harvest pro- This approach can re-create the in vivo tissue environment
cess. The PDMS membrane templates to encapsulate cells to mimic the in vivo cell –matrix interactions.
for forming MCSs have been developed [113,114]. The col- Hardelauf et al. [116] designed an array of PDMS micro-
lagen hydrogel mixed with NIH 3T3 cells and the wells to produce uniform tumour spheroids. Human colon
4 h post-seeding 12 h post-seeding 24 h post-seeding 48 h post-seeding 9

(a)

rsif.royalsocietypublishing.org
J. R. Soc. Interface 14: 20160877
(b)

Figure 5. Adipose stem cells (ASCs) cell– matrix interactions in poly(L-glutamic acid)/CS scaffold [73]. (a) Cells were stained with green fluorescent dye. The aggre-
gate formation at the first 48 h within the scaffold was observed. After 48 h, MCS size achieved was around 80 – 110 mm. (b) SEM images indicated the
multicellular aggregation progression and morphology change. Images from 12 to 48 h showed three steps of MCSs formation process. At 12 h cell-to-cell contact
initiated, followed by cadherin expression and accumulation, then ECM deposition occurred, and the compact spheroids structure was obtained without distinguished
sing cell morphology. Scale bar is 100 mm for (a) and 50 mm for (b). (Online version in colour.)

carcinoma cells (HT29), BT474 breast carcinoma cells and removal of waste droplets. Lee et al. [121] proposed an
NCI-H1792 lung carcinoma cells were pumped into the approach for in situ MCSs formation and encapsulation.
PDMS microwells and MCSs were observed after 3–4 days. They used a PDMS mould to form uniform-sized HepG2
The microwells were further modified to be concave in the MCSs within the alginate hydrogel scaffold in the concave
bottom to culture hepatoshpere and hepG2 spheroids for wells, and the nano-porous membrane to control the diffu-
drug screening [117]. Gong et al. [118] used the same concave sion of cross-linker calcium ions for alginate gelation.
microwell approach but replaced PDMS with agarose micro- Instead of using a PDMS module or template, Zhao et al.
plates to culture MCF-7 spheroids to test cancer drugs. [122] managed to employ patterned non-adhesive poly(2-
Instead of a concave well at the bottom, a side-chamber hydroxyethyl methacrylate) hydrogel films in the cell culture
design was used to culture MCSs [107]. In this device, plates to fabricate uniform-sized MCSs. The swelling-induced
two-layer PDMS microchannels were separated by a wrinkling of the film created a pattern composed of uniform
semi-permeable polycarbonate membrane. Twenty-eight concaves and cells were accumulated in the centre of the con-
dead-end side-chambers in the upper channel were designed caves to self-assemble into MCSs. Different from other
to capture and stabilize PC-3 prostate cancer cells, meanwhile microfabrication approaches, the hydrogel film is simple to
the lower channel allowed continuous medium flow for be fabricated, and spheroids are much easier to be produced
nutrient and waste diffusion. This design kept the spheroids in the film.
stationary during media exchange and MCSs were monitored A portable bioreactor was developed to maintain a ster-
in situ during long-term cultures. Beside concave microwells, ile microenvironment and sustain cell growth, maturation
other similar designs for MCS culture have been proposed. and organ formation [111]. MCF-AT1 cells were encapsu-
Jin et al. [119] proposed a design to remove cell trapping bar- lated into the Matrigel that is placed at the bottom part of
riers to facilitate MCS harvest. Cell suspension was supplied the PDMS bioreactor. Nutrients were perfused through
via the cell inlet port and distributed into four culture wells. the top section of the bioreactor. MCF-AT1 MCSs were
Pressure from the membrane pressure port deformed the harvested after couple of days. This bioreactor maintained
membrane at the bottom of the culture wells to form a horse- cell viability for long-term culture, and also allowed visual-
shoe shape for trapping cells inside the wells. After spheroid ization of the tumour spheroid formation progress through
formation, the membrane was deflated by reducing the the transparent PDMS.
pressure. Cell trapping barriers were removed and spheroids
were harvested at the spheroid collection port. Kim et al. [120]
developed a three-dimensional tumour spheroid chip with
5.2. Micro-droplets-based multicellular spheroid
balanced droplet dispensing. The hydraulic-head difference formation
between the nutrient stream inlet and the waste outlet trig- Uniform-size droplets generated from microfluidic devices pro-
gered the removal of waste droplets at the outlet. A fresh vide a confined environment for cells and hydrogels inside the
medium droplet was supplied by the dispensing layer due droplets offer a three-dimensional physical support for for-
to the decreased pressure caused by volume expansion. mation of functional MCSs. There are two configurations of
This design allowed automatic supply of fresh medium and microfluidic devices: flow focusing and T-junction are often
(a) cell and pre-gel (b) 10

rsif.royalsocietypublishing.org
micro-mould

gelatione of polymer

J. R. Soc. Interface 14: 20160877


(c)
water
phase with
(1) cells (2)

oil
phase

(3) (4)

0 min 150 min

Figure 6. Microfluidic methods for MCS fabrication. (a) MCS formation in micro moulds. (b) PC-3 prostate cancer MCSs formation in microwells [107]. (c) MCS
formation in droplets generated from microfluidic channels: (1) T-junction cell encapsulation, (2) flow-focusing cell encapsulation, (3) double emulsion cell encap-
sulation and (4) human mesenchymal stem cells MCSs formation inside droplets [133]. (Online version in colour.)

employed to generate uniform-size droplets. In the flow-focus- formed MCSs encapsulated inside the hydrogel droplets are
ing device shown in figure 5, the aqueous phase with cells injected into the patient’s body, they are protected from
through the middle inlet as a dispersed fluid, and the immiscible host immune responses, which minimizes administration of
organic solvent flows through both side inlets as a continuous immunosuppressive drugs and increases the successful trans-
fluid. The shear force from the continuous fluid squeezes the dis- plantation rate [125]. This approach also has a high degree of
persed fluid to generate spherical individual droplets. On control over the morphological and physical properties [109].
the other hand, in the T-junction device, the dispersed fluid Sakai et al. [126] used flow focusing microdevices to gener-
penetrates the continuous fluid to form droplets and the ate droplets for encapsulating HepG2 cells (figure 5c). Gelatin
newly formed droplets are swept by the continuous fluid. The hydrogel with cells flew through from the top inlet, and paraf-
hydrogels inside the droplets are often gelled due to external fin oil flew through the left inlet. The hydrogel-containing
stimuli, such as temperature, light or ions. Thermal responsive droplets were heated to the melt point and the gelation was
hydrogels, such as agarose, NIPAM-based hydrogels or gelatin, realized by cooling down to the gelation point when collecting
solidify due to a temperature shift of the hydrogel system. Photo- the droplets. The size of the droplets from 300 to 100 mm was
resistive hydrogels, such as PEG, form the physical gel by cross- controlled by varying the paraffin flow rate. The HepG2 cells
linking triggered by exposure to UV light. Alginate is solidified were cultured inside the droplets and they were aggregated
by the introduction of divalent ion into the system (e.g. Ca2þ). to form MCSs in 24 h. This approach generated the desired
Normally, UV exposure or dramatic temperature change size of MCSs since the size of droplet was controllable. Yoon
has a negative impact on cell viability as well as its physiology. et al. [127] proposed a similar approach, but they used alginate
Hence, the ion-based cross-linking gelation method is con- instead of gelatin to minimize the damage in the gelation pro-
sidered a safer alternative for cell encapsulation. cess on the cell viability. Magnetic iron oxide nanoparticles
This droplet-based method is rapid and high throughput. were uptaken with the spheroids to achieve easy collection
Velasco et al. [123] demonstrated that cell-loaded hydrogel and separation of the spheroids during the harvest process.
droplets significantly reduced the intensive labour needed Instead of using alginate or gelatin, a mixture of alginate and
to fabricate MCSs. In the porous micro droplets, oxygen Matrigel was introduced for MCS culture by Wang et al.
and nutrients were diffused in and metabolic waste diffused [105]. This mixture gels performed better in the formation of
out to maximize the cell viability [124]. In addition, when the HeLa spheroids compared with pure alginate beads. Tsuda
et al. [128] used self-assembling peptides (SAP, Purmatrix expansion while in vivo tumour spheroids form from a rela- 11
RADA 16) to encapsulate bovine carotid artery endothelial tively low cell density and the size of the tumour spheroids

rsif.royalsocietypublishing.org
cells [129,130]. This synthetic peptide was functionalized increases due to cell expansion inside the structure. Secondly,
with other monomers to enhance cell attachment, increase the reproducibility and quality assurance of conventional
cell proliferation and promote cell differentiation. The peptide means are low. Thirdly, the microenvironment and marcoen-
hydrogels inside the droplets were solidified by exposure to vironment for MCS formation from current methods are still
the cross-linking agent in the continuous phase. After cultur- different from in vivo, which results in different cellular beha-
ing the cells inside the droplets for 3 days, endothelial cells viours. For instance, cell migration between MCSs is not
inside droplets migrated to form loose aggregates due to initial realized from current methods. There is a gap in producing
cell-to-cell contacts, spheroids were formed in long-term functional tissues through the in vitro culture due to lack of
culture. The PuraMatrix hydrogels was solidified in the heterogeneous cell –cell interactions, cell– ECM organization
presence of certain ion concentrations. This hydrogel was and cellular signalling pathways within the tissue. Hence,

J. R. Soc. Interface 14: 20160877


attempted to encapsulate HepG2 cells in a double T-junction the correlation between real tissues and MCSs should be
and the gelation was achieved by cross-linking with the ions thoroughly documented through gene expression profiling
in the cell medium [131]. Thermo-responsive microgel poly analysis. Moreover, cell behaviours may alter when they are
(N-Isopropylacrylamide-co-acrylic acid) (P(NIAM-AA)) dro- exposed to different microenvironments/marcoenvironments
plets were also used to encapsulate and culture HeLa cell and cells are able to adapt themselves for different types of
spheroids [132]. The porous structure of the hydrogel allowed MCSs in a real tissue, which are still missing in the in vitro
sufficient mass transportation including nutrients, oxygen and MCS model. Furthermore, the heterogeneous metabolism
bio-wastes, which was beneficial for long-term culture. In and gene expression in MCSs can be influenced by the
addition, the thermal reversibility of this hydrogel facilitated MCS size. Thus, a uniform-size of MCSs should be pursued.
simple recovery of MCSs easily after long-term culture without The methods for fabrication of MCSs in vitro have been
introduction of toxic chemicals. significantly developed in the past decade. The integration
Chan et al. [133] developed a double emulsion system to of microfluidics with scaffolds can be considered a promising
fabricate MCSs from bone marrow-derived human mesenchy- new approach as it can not only produce controlled uniform-
mal stem cells (hMSCs). The double emulsion was performed size MCSs, but also restore the complex cell– matrix/cell –cell
in two flow-focusing devices. The water/oil emulsion was interactions which are vital for MCSs morphology and func-
generated in the first device, which had an inner core contain- tionality. The physical matrix support provided by the
ing a mixture of hMSCs and the culture medium and an out scaffold can facilitate MCSs in developing their own extra-
layer of oil. The water/oil emulsion went through the second cellular matrices that are crucial for cell functions. The
device to generate the water/oil/water emulsion. The chambers or droplets from the microfluidics allow fabrication
middle oil layer served as a barrier to prevent the inner core of MCSs in a confined space to develop controllable uniform-
from mixing with the culture medium at the outer layer. The sized MCSs. This approach is also able to tune the microen-
selective permeability of the oil layer allowed nutrient diffu- vironment for MCS formation and growth to mimic the in
sion from the outer aqueous phase into the inner core and vivo conditions. In addition, the potential of commercializa-
waste removal in the opposite direction. tion makes the approach even more attractive. More elegant
designs for microfluidics and scaffolds for MCSs formation
are needed to create physiological relevant microenviron-
6. Discussion and the future ments and marcoenvironments to address the above
challenges for MCSs formation.
With the advances in MCSs culture, the in vitro model is able
to mimic the in vivo characteristics in many aspects. The MCS
complex structure allows a better understanding of cell –cell Authors’ contributions. X.C. organized, prepared and wrote this paper;
Y.H. helped to organize figures and tables; H.Z. revised the
and cell– matrix interactions. In addition, recapitulation of
manuscript.
the in vivo microenvironment by MCSs enables fundamental
Competing interests. We declare we have no competing interests.
research on cancer biology and tissue development and pro-
Funding. H.Z. would like to acknowledge the financial support from
vides an opportunity for culturing functional tissues in vitro. ARC Discovery Project (DP160104632) and the Medical Advance-
However, there are a few challenges for fabricating MCSs. ment Without Animal (MAWA) Trust.
Firstly, a high initial cell density is required to form spheroids Acknowledgement. X.C. would like to acknowledge the University of
and cells in the MCSs structure are not required for further Adelaide, Faculty of ECMS, for providing a scholarship.

References
1. Nath S, Devi GR. 2016 Three-dimensional influence of biomaterial chemistry and tenascin-C. 4. Achilli T-M, Meyer J, Morgan JR. 2012 Advances
culture systems in cancer research: focus on Exp. Cell Res. 301, 179–188. (doi:10.1016/j.yexcr. in the formation, use and understanding of
tumor spheroid model. Pharmacol. Ther. 2004.07.027) multi-cellular spheroids. Expert Opin Biol.
163, 94 –108. (doi:10.1016/j.pharmthera. 3. Mehta G, Hsiao AY, Ingram M, Luker GD, Takayama Ther. 12, 1347– 1360. (doi:10.1517/14712598.
2016.03.013) S. 2012 Opportunities and challenges for use of 2012.707181)
2. Mahmood TA, de Jong R, Riesle J, Langer R, van tumor spheroids as models to test drug delivery and 5. Moscona A, Moscona H. 1952 The dissociation and
Blitterswijk CA. 2004 Adhesion-mediated signal efficacy. J. Control Release. 164, 192– 204. (doi:10. aggregation of cells from organ rudiments of the
transduction in human articular chondrocytes: the 1016/j.jconrel.2012.04.045) early chick embryo. J. Anat. 86, 287–301.
6. Wu Y, Zhao Z, Guan Y, Zhang Y. 2014 Galactosylated 21. Alhasan L, Qi A, Al-Abboodi A, Rezk A, Chan PPY, 34. Korff T, Augustin HG. 1998 Integration of 12
reversible hydrogels as scaffold for HepG2 spheroid Iliescu C, Yeo LY. 2016 Rapid enhancement of endothelial cells in multicellular spheroids prevents

rsif.royalsocietypublishing.org
generation. Acta Biomater. 10, 1965– 1974. (doi:10. cellular spheroid assembly by acoustically driven apoptosis and induces differentiation. J. Cell Biol.
1016/j.actbio.2013.12.044) microcentrifugation. ACS Biomater. Sci. Eng. 2, 143, 1341– 1352. (doi:10.1083/jcb.143.5.1341)
7. Lei Y, Schaffer DV. 2013 A fully defined and scalable 1013 –1022. (doi:10.1021/acsbiomaterials.6b00144) 35. Debnath J, Collins NL, Mills KR, Reginato MJ,
3D culture system for human pluripotent stem cell 22. Tzanakakis ES, Hansen LK, Hu W-S. 2001 The role of Muthuswamy SK. 2002 The role of apoptosis in
expansion and differentiation. Proc. Natl Acad. Sci. actin filaments and microtubules in hepatocyte creating and maintaining luminal space within
USA 110, E5039– E5048. (doi:10.1073/pnas. spheroid self-assembly. Cell Motil. Cytoskeleton. 48, normal and oncogene-expressing mammary acini.
1309408110) 175 –189. (doi:10.1002/1097-0169(200103)48: Cell 111, 29 –40. (doi:10.1016/S0092-
8. Vinci M et al. 2012 Advances in establishment and 3,175::AID-CM1007.3.0.CO;2-2) 8674(02)01001-2)
analysis of three-dimensional tumor spheroid-based 23. Kimlin LC, Casagrande G, Virador VM. 2013 In vitro 36. Weaver VM, Lelievre S, Lakins JN, Chrenek MA,
functional assays for target validation and drug three-dimensional (3D) models in cancer research: Jones JCR, Giancotti F, Werb Z, Bissell MJ. 2002 b4

J. R. Soc. Interface 14: 20160877


evaluation. BMC Biol. 10, 1 –21. (doi:10.1186/1741- an update. Mol. Carcinog. 52, 167–182. (doi:10. integrin-dependent formation of polarized three-
7007-10-29) 1002/mc.21844) dimensional architecture confers resistance to
9. Abu-Absi SF, Friend JR, Hansen LK, Hu W-S. 2002 24. Kim JB, Stein R, O’Hare MJ. 2004 Three-dimensional apoptosis in normal and malignant mammary
Structural polarity and functional bile canaliculi in in vitro tissue culture models of breast cancer: a epithelium. Cancer Cell. 2, 205–216. (doi:10.1016/
rat hepatocyte spheroids. Exp. Cell Res. 274, 56– 67. review. Breast Cancer Res. Treat. 85, 281 –291. S1535-6108(02)00125-3)
(doi:10.1006/excr.2001.5467) (doi:10.1023/B:BREA.0000025418.88785.2b) 37. Takezawa T, Yamazaki M, Mori Y, Yonaha T,
10. Kelm JM, Djonov V, Ittner LM, Fluri D, Born W, 25. Ronnov-Jessen L, Petersen OW, Koteliansky VE, Bissell Yoshizato K. 1992 Morphological and immuno-
Hoerstrup SP, Fussenegger M. 2006 Design of custom- MJ. 1995 The origin of the myofibroblasts in breast cytochemical characterization of a hetero-spheroid
shaped vascularized tissues using microtissue cancer. Recapitulation of tumor environment in culture composed of fibroblasts and hepatocytes. J. Cell Sci.
spheroids as minimal building units. Tissue Eng. 12, unravels diversity and implicates converted fibroblasts 101, 495–501.
2151 –2160. (doi:10.1089/ten.2006.12.2151) and recruited smooth muscle cells. J. Clin. Invest. 95, 38. Lu H-F, Chua K-N, Zhang P-C, Lim W-S,
11. Mironov V, Visconti RP, Kasyanov V, Forgacs G, Drake 859–873. (doi:10.1172/JCI117736) Ramakrishna S, Leong KW, Mao H-Q. 2005 Three-
CJ, Markwald RR. 2009 Organ printing: tissue 26. Chen Y, Stump RJ, Lovicu FJ, Shimono A, McAvoy dimensional co-culture of rat hepatocyte spheroids
spheroids as building blocks. Biomaterials 30, 2164 – JW. 2008 Wnt signaling is required for organization and NIH/3T3 fibroblasts enhances hepatocyte
2174. (doi:10.1016/j.biomaterials.2008.12.084) of the lens fiber cell cytoskeleton and development functional maintenance. Acta Biomater. 1,
12. Lee BH, Kim MH, Lee JH, Seliktar D, Cho N-J, Tan of lens three-dimensional architecture. Dev. Biol. 399–410. (doi:10.1016/j.actbio.2005.04.003)
LP. 2015 Modulation of Huh7.5 spheroid formation 324, 161 –176. (doi:10.1016/j.ydbio.2008.09.002) 39. Lee KW et al. 2004 Influence of pancreatic islets on
and functionality using modified peg-based 27. Han M, Oba M, Nishiyama N, Kano MR, Kizaka- spheroid formation and functions of hepatocytes in
hydrogels of different stiffness. PLoS ONE 10, Kondoh S, Kataoka K. 2009 Enhanced percolation hepatocyte-pancreatic islet spheroid culture. Tissue
e0118123. (doi:10.1371/journal.pone.0118123) and gene expression in tumor hypoxia by PEGylated Eng. 10, 965 –977.
13. Lin R-Z, Chou L-F, Chien C-CM, Chang H-Y. 2006 polyplex micelles. Mol. Ther. 17, 1404–1410. 40. Murakami S, Ijima H, Ono T, Kawakami K. 2004
Dynamic analysis of hepatoma spheroid formation: (doi:10.1038/mt.2009.119) Development of co-culture system of hepatocytes
roles of E-cadherin and b1-integrin. Cell Tissue Res. 28. Santini MT, Rainaldi G, Indovina PL. 2000 Apoptosis, with bone marrow cells for expression and
324, 411–422. (doi:10.1007/s00441-005-0148-2) cell adhesion and the extracellular matrix in the maintenance of hepatic functions. Int. J. Artif.
14. Robinson EE, Foty RA, Corbett SA. 2004 Fibronectin three-dimensional growth of multicellular tumor Organs. 27, 118–126.
matrix assembly regulates a5b1-mediated cell spheroids. Crit. Rev. Oncol. Hematol. 36, 75 –87. 41. Lee GY, Kenny PA, Lee EH, Bissell MJ. 2007 Three-
cohesion. Mol. Biol. Cell. 15, 973–981. (doi:10. (doi:10.1016/S1040-8428(00)00078-0) dimensional culture models of normal and
1091/mbc.E03-07-0528) 29. Khademhosseini A, Eng G, Yeh J, Fukuda J, Blumling malignant breast epithelial cells. Nat. Methods. 4,
15. Ivascu A, Kubbies M. 2007 Diversity of cell-mediated III J, Langer R, Burdick JA. 2006 Micromolding of 359–365. (doi:10.1038/nmeth1015)
adhesions in breast cancer spheroids. Int. J. Oncol. photocrosslinkable hyaluronic acid for cell 42. Gottfried E, Kunz-Schughart LA, Andreesen R, Kreutz
31, 1403 –1413. encapsulation and entrapment. J. Biomed. Mater. Res. M. 2006 Brave little world: spheroids as an in vitro
16. Shimazui T et al. 2004 Role of complex cadherins in A. 79, 522–532. (doi:10.1002/jbm.a.30821) model to study tumor-immune-cell interactions. Cell
cell-cell adhesion evaluated by spheroid formation 30. Curcio E, Salerno S, Barbieri G, De Bartolo L, Drioli E, Cycle 5, 691–695. (doi:10.4161/cc.5.7.2624)
in renal cell carcinoma cell lines. Oncol. Rep. 11, Bader A. 2007 Mass transfer and metabolic reactions in 43. Seidl P, Huettinger R, Knuechel R, Kunz-Schughart
357–360. hepatocyte spheroids cultured in rotating wall gas- LA. 2002 Three-dimensional fibroblast–tumor cell
17. Cavallaro U, Christofori G. 2004 Cell adhesion and permeable membrane system. Biomaterials 28, interaction causes downregulation of RACK1 mRNA
signalling by cadherins and Ig-CAMs in cancer. Nat. 5487–5497. (doi:10.1016/j.biomaterials.2007.08.033) expression in breast cancer cells in vitro. Int. J. Cancer.
Rev. Cancer. 4, 118– 132. (doi:10.1038/nrc1276) 31. Mueller-Klieser W. 1984 Method for the 102, 129–136. (doi:10.1002/ijc.10675)
18. Ivascu A, Kubbies M. 2006 Rapid generation of determination of oxygen consumption rates and 44. Kunz-Schughart LA, Heyder P, Schroeder J, Knuechel
single-tumor spheroids for high-throughput cell diffusion coefficients in multicellular spheroids. R. 2001 A heterologous 3-D coculture model of
function and toxicity analysis. J. Biomol. Screen. 11, Biophys. J. 46, 343 –348. (doi:10.1016/S0006- breast tumor cells and fibroblasts to study tumor-
922–932. (doi:10.1177/1087057106292763) 3495(84)84030-8) associated fibroblast differentiation. Exp. Cell Res.
19. Panchina Y, Kelmanson I, Matz M, Lukyanov K, Usman 32. Alvarez-Pérez J, Ballesteros P, Cerdán S. 2005 266, 74– 86. (doi:10.1006/excr.2001.5210)
N, Lukyanov S. 2000 A ubiquitous family of putative Microscopic images of intraspheroidal pH by 1H 45. Kim JB. 2005 Three-dimensional tissue culture
gap junction molecules. Curr. Biol. 10, R473 –R4R4. magnetic resonance chemical shift imaging of pH models in cancer biology. Semin. Cancer Biol. 15,
(doi:10.1016/S0960-9822(00)00576-5) sensitive indicators. Magn. Reson. Mater. Phys. Biol. 365–377. (doi:10.1016/j.semcancer.2005.05.002)
20. Bao BA, Lai CP, Naus CC, Morgan JR. 2012 Med. 18, 293–301. (doi:10.1007/s10334-005-0013-z) 46. Qihao Z, Xigu C, Guanghui C, Weiwei Z. 2007
Pannexin1 drives multicellular aggregate 33. Carlsson J, Acker H. 1988 Relations between pH, spheroid formation and differentiation into
compaction via a signaling cascade that remodels oxygen partial pressure and growth in cultured cell hepatocyte-like cells of rat mesenchymal stem cell
the actin cytoskeleton. J. Biol. Chem. 287, 8407 – spheroids. Int. J. Cancer. 42, 715 –720. (doi:10. induced by co-culture with liver cells. DNA Cell Biol.
8416. (doi:10.1074/jbc.M111.306522) 1002/ijc.2910420515) 26, 497 –503. (doi:10.1089/dna.2006.0562)
47. Ronnov-Jessen L, Petersen OW, Bissell MJ. 1996 mammalian cells in multilayer aggregates using panel of three-dimensional models for studies of 13
Cellular changes involved in conversion of normal to dielectrophoresis. Biotechnol. Bioeng. 98, 694–700. prostate cancer growth, invasion and drug

rsif.royalsocietypublishing.org
malignant breast: importance of the stromal (doi:10.1002/bit.21416) responses. PLoS ONE 5, e10431. (doi:10.1371/
reaction. Physiol. Rev. 76, 69 –125. 62. Chen K et al. 2016 Rapid formation of size- journal.pone.0010431)
48. Martin GR. 1980 Teratocarcinomas and mammalian controllable multicellular spheroids via 3D acoustic 75. Dubiak-Szepietowska M, Karczmarczyk A, Jönsson-
embryogenesis. Science 209, 768–776. (doi:10. tweezers. Lab. Chip. 16, 2636–2643. (doi:10.1039/ Niedziółka M, Winckler T, Feller K-H. 2016
1126/science.6250214) C6LC00444J) Development of complex-shaped liver multicellular
49. Fischbach C, Chen R, Matsumoto T, Schmelzle T, 63. Lin RZ, Chang HY. 2008 Recent advances in three- spheroids as a human-based model for nanoparticle
Brugge JS, Polverini PJ, Mooney DJ. 2007 dimensional multicellular spheroid culture for toxicity assessment in vitro. Toxicol. Appl. Pharmacol.
Engineering tumors with 3D scaffolds. Nat. Methods biomedical research. Biotechnol. J. 3, 1172–1184. 294, 78–85. (doi:10.1016/j.taap.2016.01.016)
4, 855–860. (doi:10.1038/nmeth1085) (doi:10.1002/biot.200700228) 76. Asghar W, El Assal R, Shafiee H, Pitteri S,
50. Ma H et al. 2012 Multicellular tumor spheroids as 64. Haycock JW. 2011 3D Cell culture: a review of Paulmurugan R, Demirci U. 2015 Engineering cancer

J. R. Soc. Interface 14: 20160877


an in vivo-like tumor model for three-dimensional current approaches and techniques. In 3D cell microenvironments for in vitro 3-D tumor models.
imaging of chemotherapeutic and nano material culture: methods and protocols (ed. JW Haycock), Mater. Today. 18, 539–553. (doi:10.1016/j.mattod.
cellular penetration. Mol. Imaging. 11, 487. pp. 1–15. Totowa, NJ: Humana Press. 2015.05.002)
51. Cui X, Dini S, Dai S, Bi J, Binder BJ, Green JEF, 65. Ruedinger F, Lavrentieva A, Blume C, Pepelanova I, 77. Salmenperä P, Karhemo P-R, Räsänen K, Laakkonen
Zhang H. 2016 A mechanistic study on tumour Scheper T. 2015 Hydrogels for 3D mammalian cell P, Vaheri A. 2016 Fibroblast spheroids as a model to
spheroid formation in thermosensitive hydrogels: culture: a starting guide for laboratory practice. study sustained fibroblast quiescence and their
experiments and mathematical modelling. RSC Adv. Appl. Microbiol. Biotechnol. 99, 623– 636. (doi:10. crosstalk with tumor cells. Exp. Cell Res. 345,
6, 73 282 –73 291. (doi:10.1039/C6RA11699J) 1007/s00253-014-6253-y) 17– 24. (doi:10.1016/j.yexcr.2016.05.005)
52. Yuhas JM, Li AP, Martinez AO, Ladman AJ. 1977 A 66. Baharvand H, Hashemi SM, Ashtiani SK, Farrokhi A. 78. Dingle Y-TL et al. 2015 Three-dimensional neural
simplified method for production and growth of 2006 Differentiation of human embryonic stem cells spheroid culture: an in vitro model for cortical
multicellular tumor spheroids. Cancer Res. 37, into hepatocytes in 2D and 3D culture systems in studies. Tissue Eng. Part C Methods 21, 1274–1283.
3639 –3643. vitro. Int. J. Dev. Biol. 50, 645. (doi:10.1387/ijdb. (doi:10.1089/ten.tec.2015.0135)
53. Kelm JM, Timmins NE, Brown CJ, Fussenegger M, 052072hb) 79. Williams SK, Touroo JS, Church KH, Hoying JB. 2013
Nielsen LK. 2003 Method for generation of 67. Holliday DL, Brouilette KT, Markert A, Gordon LA, Encapsulation of adipose stromal vascular fraction cells
homogeneous multicellular tumor spheroids Jones JL. 2009 Novel multicellular organotypic in alginate hydrogel spheroids using a direct-write
applicable to a wide variety of cell types. Biotechnol. models of normal and malignant breast: tools for three-dimensional printing system. BioRes. Open
Bioeng. 83, 173– 180. (doi:10.1002/bit.10655) dissecting the role of the microenvironment in Access 2, 448–454. (doi:10.1089/biores.2013.0046)
54. Oliveira MB, Neto AI, Correia CR, Rial-Hermida MI, breast cancer progression. Breast Cancer Res. 11, 80. Shakibaei M, Kraehe P, Popper B, Shayan P, Goel A,
Alvarez-Lorenzo C, Mano JF. 2014 Superhydrophobic 1 –11. (doi:10.1186/bcr2218) Buhrmann C. 2015 Curcumin potentiates antitumor
chips for cell spheroids high-throughput generation 68. Fang JY, Tan S-J, Yang Z, Tayag C, Han B. 2014 activity of 5-fluorouracil in a 3D alginate tumor
and drug screening. ACS Appl. Mater. Interfaces 6, Tumor bioengineering using a transglutaminase microenvironment of colorectal cancer. BMC Cancer
9488–9495. (doi:10.1021/am5018607) crosslinked hydrogel. PLoS ONE 9, e105616. (doi:10. 15, 1 –15. (doi:10.1186/s12885-015-1291-0)
55. Kim JB. (ed.) 2005 Three-dimensional tissue culture 1371/journal.pone.0105616) 81. Murphy KC, Fang SY, Leach JK. 2014 Human
models in cancer biology. Seminars in cancer biology. 69. Gurski LA, Jha AK, Zhang C, Jia X, Farach-Carson MC. mesenchymal stem cell spheroids in fibrin hydrogels
Amsterdam, The Netherlands: Elsevier. 2009 Hyaluronic acid-based hydrogels as 3D exhibit improved cell survival and potential for bone
56. Sutherland RM, Inch WR, McCredie JA, Kruuv J. matrices for in vitro evaluation of chemotherapeutic healing. Cell Tissue Res. 357, 91 –99. (doi:10.1007/
1970 A multi-component radiation survival curve drugs using poorly adherent prostate cancer cells. s00441-014-1830-z)
using an in vitro tumour model. Int. J. Radiat. Biol. Biomaterials 30, 6076–6085. (doi:10.1016/j. 82. Liu J et al. 2012 Soft fibrin gels promote selection
Related Stud. Phys. Chem. Med. 18, 491–495. biomaterials.2009.07.054) and growth of tumorigenic cells. Nat. Mater. 11,
(doi:10.1080/09553007014551401) 70. Ananthanarayanan B, Kim Y, Kumar S. 2011 734–741. (doi:10.1038/nmat3361)
57. Ingram M, Techy GB, Saroufeem R, Yazan O, Elucidating the mechanobiology of malignant brain 83. Shubin AD, Felong TJ, Graunke D, Ovitt CE, Benoit
Narayan KS, Goodwin TJ, Spaulding GF. 1997 tumors using a brain matrix-mimetic hyaluronic acid DSW. 2015 Development of poly(ethylene glycol)
Three-dimensional growth patterns of various hydrogel platform. Biomaterials 32, 7913–7923. hydrogels for salivary gland tissue engineering
human tumor cell lines in simulated microgravity (doi:10.1016/j.biomaterials.2011.07.005) applications. Tissue Eng. Part A. 21, 1733–1751.
of a NASA bioreactor. In Vitro Cell. Dev. Biol. 71. Liu Y, Shu XZ, Prestwich GD. 2007 Tumor (doi:10.1089/ten.tea.2014.0674)
Anim. 33, 459 –466. (doi:10.1007/s11626-997- engineering: orthotopic cancer models in mice 84. Chou M-J, Hsieh C-H, Yeh P-L, Chen P-C, Wang C-H,
0064-8) using cell-loaded, injectable, cross-linked Huang Y-Y. 2013 Application of open porous
58. Ino K, Ito A, Honda H. 2007 Cell patterning using hyaluronan-derived hydrogels. Tissue Eng. 13, poly(D,L-lactide-co-glycolide) microspheres and the
magnetite nanoparticles and magnetic force. 1091 –1101. (doi:10.1089/ten.2006.0297) strategy of hydrophobic seeding in hepatic tissue
Biotechnol. Bioeng. 97, 1309 –1317. (doi:10.1002/ 72. Burdett E, Kasper FK, Mikos AG, Ludwig JA. 2010 cultivation. J. Biomed. Mater. Res. A. 101, 2862–
bit.21322) Engineering tumors: a tissue engineering 2869. (doi:10.1002/jbm.a.34594)
59. Bae J-E et al. 2011 The effect of static magnetic perspective in cancer biology. Tissue Eng. 85. Wang D, Cheng D, Guan Y, Zhang Y. 2011
fields on the aggregation and cytotoxicity of Part B Rev. 16, 351 –359. (doi:10.1089/ten.teb. Thermoreversible hydrogel for in situ generation
magnetic nanoparticles. Biomaterials 32, 9401 – 2009.0676) and release of HepG2 spheroids. Biomacromolecules
9414. (doi:10.1016/j.biomaterials.2011.08.075) 73. Zhang K, Yan S, Li G, Cui L, Yin J. 2015 In-situ birth 12, 578 –584. (doi:10.1021/bm101187b)
60. Sebastian A, Buckle A-M, Markx GH. 2006 Formation of MSCs multicellular spheroids in poly(l-glutamic 86. Fong ELS et al. 2013 Modeling Ewing sarcoma
of multilayer aggregates of mammalian cells by acid)/chitosan scaffold for hyaline-like cartilage tumors in vitro with 3D scaffolds. Proc. Natl Acad.
dielectrophoresis. J. Micromech. Microeng. 16, 1769. regeneration. Biomaterials 71, 24 –34. (doi:10. Sci. USA 110, 6500–6505. (doi:10.1073/pnas.
(doi:10.1088/0960-1317/16/9/003) 1016/j.biomaterials.2015.08.037) 1221403110)
61. Sebastian A, Buckle AM, Markx GH. 2007 Tissue 74. Härmä V, Virtanen J, Mäkelä R, Happonen A, Mpindi 87. Talukdar S, Kundu SC. 2012 A non-mulberry silk fibroin
engineering with electric fields: Immobilization of J-P, Knuuttila M, Nees M. 2010 A comprehensive protein based 3d in vitro tumor model for evaluation of
anticancer drug activity. Adv. Funct. Mater. 22, breast cancer bone metastasis. PLoS ONE 5, e8849. macroscopic 3D tissue architecture. Adv. Mater. 23, 14
4778–4788. (doi:10.1002/adfm.201200375) (doi:10.1371/journal.pone.0008849) H90–H94. (doi:10.1002/adma.201004375)

rsif.royalsocietypublishing.org
88. Nyga A, Cheema U, Loizidou M. 2011 3D tumour 102. Gu J, Zhao Y, Guan Y, Zhang Y. 2015 Effect of 116. Hardelauf H et al. 2011 Microarrays for the scalable
models: novel in vitro approaches to cancer studies. particle size in a colloidal hydrogel scaffold for 3D production of metabolically relevant tumour
J. Cell Commun. Signal. 5, 239 –248. (doi:10.1007/ cell culture. Colloids Surf. B. 136, 1139– 1147. spheroids: a tool for modulating chemosensitivity
s12079-011-0132-4) (doi:10.1016/j.colsurfb.2015.11.021) traits. Lab. Chip. 11, 419–428. (doi:10.1039/
89. Walters BD, Stegemann JP. 2014 Strategies for 103. Liu Y, Guan Y, Zhang Y. 2015 Chitosan as inter- C0LC00089B)
directing the structure and function of three- cellular linker to accelerate multicellular spheroid 117. Wong SF, No da Y, Choi YY, Kim DS, Chung BG, Lee
dimensional collagen biomaterials across length generation in hydrogel scaffold. Polymer 77, SH. 2011 Concave microwell based size-controllable
scales. Acta Biomater. 10, 1488 –1501. (doi:10. 366 –376. (doi:10.1016/j.polymer.2015.09.073) hepatosphere as a three-dimensional liver tissue
1016/j.actbio.2013.08.038) 104. Gao D, Liu H, Jiang Y, Lin J-M, Gao D, Liu H, Jiang model. Biomaterials 32, 8087–8096. (doi:10.1016/
90. Dagalakis N, Flink J, Stasikelis P, Burke J, Yannas I. Y. 2012 Recent developments in microfluidic devices j.biomaterials.2011.07.028)

J. R. Soc. Interface 14: 20160877


1980 Design of an artificial skin. III. Control of pore for in vitro cell culture for cell-biology research. 118. Gong X, Lin C, Cheng J, Su J, Zhao H, Liu T, Wen X,
structure. J. Biomed. Mater. Res. 14, 511 –528. TrAC, Trends Anal. Chem.. 35, 150 –164. (doi:10. Zhao P, Kerkis I. 2015 Generation of multicellular
(doi:10.1002/jbm.820140417) 1016/j.trac.2012.02.008) tumor spheroids with microwell-based agarose
91. Themistocleous G et al. 2004 Three-dimensional 105. Wang Y, Wang J. 2014 Mixed hydrogel bead-based scaffolds for drug testing. PLoS ONE 10, e0130348.
type I collagen cell culture systems for the study of tumor spheroid formation and anticancer drug (doi:10.1371/journal.pone.0130348)
bone pathophysiology. In Vivo 18, 687–696. testing. Analyst 139, 2449–2458. (doi:10.1039/ 119. Jin HJ, Cho YH, Gu JM, Kim J, Oh YS. 2011 A
92. Yamada M, Hori A, Sugaya S, Yajima Y, Utoh R, C4AN00015C) multicellular spheroid formation and extraction chip
Yamato M, Seki M. 2015 Cell-sized condensed 106. Anada T, Fukuda J, Sai Y, Suzuki O. 2012 An using removable cell trapping barriers. Lab. Chip.
collagen microparticles for preparing oxygen-permeable spheroid culture system for the 11, 115 –119. (doi:10.1039/C0LC00134A)
microengineered composite spheroids of primary prevention of central hypoxia and necrosis of 120. Kim T, Doh I, Cho Y-H. 2012 A 3D tumor spheroid
hepatocytes. Lab. Chip. 15, 3941 –3951. (doi:10. spheroids. Biomaterials 33, 8430– 8441. (doi:10. chip with the pharmacokinetic drug elimination
1039/C5LC00785B) 1016/j.biomaterials.2012.08.040) model developed by balanced droplet dispensing.
93. Xu F, Burg KJ. 2007 Three-dimensional 107. Hsiao AY, Torisawa Y-S, Tung Y-C, Sud S, Taichman Sens. Actuators B 174, 436– 440. (doi:10.1016/j.
polymeric systems for cancer cell studies. RS, Pienta KJ, Takayama S. 2009 Microfluidic system snb.2012.08.041)
Cytotechnology 54, 135–143. (doi:10.1007/s10616- for formation of PC-3 prostate cancer co-culture 121. Lee KH, No da Y, Kim SH, Ryoo JH, Wong SF, Lee
007-9065-4) spheroids. Biomaterials 30, 3020– 3027. (doi:10. SH. 2011 Diffusion-mediated in situ alginate
94. Wang K, Kievit FM, Florczyk SJ, Stephen ZR, Zhang 1016/j.biomaterials.2009.02.047) encapsulation of cell spheroids using
M. 2015 3D porous chitosan –alginate scaffolds as 108. Montanez-Sauri SI, Beebe DJ, Sung KE. 2015 microscale concave well and nanoporous membrane.
an in vitro model for evaluating nanoparticle- Microscale screening systems for 3D cellular Lab. Chip. 11, 1168–1173. (doi:10.1039/
mediated tumor targeting and gene delivery to microenvironments: platforms, advances, and c0lc00540a)
prostate cancer. Biomacromolecules 16, 3362– 3372. challenges. Cell. Mol. Life Sci. 72, 237–249. (doi:10. 122. Zhao Z, Gu J, Zhao Y, Guan Y, Zhu XX, Zhang Y.
(doi:10.1021/acs.biomac.5b01032) 1007/s00018-014-1738-5) 2014 Hydrogel thin film with swelling-induced
95. Muzzarelli RA. 2011 Chitosan composites with 109. Kang A, Park J, Ju J, Jeong GS, Lee SH. 2014 wrinkling patterns for high-throughput generation
inorganics, morphogenetic proteins and stem cells, for Cell encapsulation via microtechnologies. of multicellular spheroids. Biomacromolecules 15,
bone regeneration. Carbohydr. Polym. 83, 1433–1445. Biomaterials 35, 2651–2663. (doi:10.1016/j. 3306– 3312. (doi:10.1021/bm500722g)
(doi:10.1016/j.carbpol.2010.10.044) biomaterials.2013.12.073) 123. Velasco D, Tumarkin E, Kumacheva E. 2012
96. Wang J-Z, Zhu Y-X, Ma H-C, Chen S-N, Chao J-Y, 110. Kwapiszewska K, Michalczuk A, Rybka M, Microfluidic encapsulation of cells in polymer
Ruan W-D, Wang D, Du F-G, Meng Y-Z. 2016 Kwapiszewski R, Brzozka Z. 2014 A microfluidic- microgels. Small 8, 1633–1642. (doi:10.1002/smll.
Developing multi-cellular tumor spheroid model based platform for tumour spheroid culture, 201102464)
(MCTS) in the chitosan/collagen/alginate (CCA) monitoring and drug screening. Lab. Chip. 14, 124. Serra M, Correia C, Malpique R, Brito C, Jensen J,
fibrous scaffold for anticancer drug screening. Mater. 2096 –2104. (doi:10.1039/C4LC00291A) Bjorquist P, Carrondo MJT, Alves PM, Chan C. 2011
Sci. Eng. C 62, 215–225. (doi:10.1016/j.msec.2016. 111. Markov DA, Lu JQ, Samson PC, Wikswo JP, Microencapsulation technology: a powerful tool for
01.045) McCawley LJ. 2012 Thick-tissue bioreactor as a integrating expansion and cryopreservation of
97. Caliari SR, Burdick JA. 2016 A practical guide to platform for long-term organotypic culture and human embryonic stem cells. PLoS ONE 6, e23212.
hydrogels for cell culture. Nat. Methods. 13, drug delivery. Lab. Chip. 12, 4560–4568. (doi:10. (doi:10.1371/journal.pone.0023212)
405–414. (doi:10.1038/nmeth.3839) 1039/c2lc40304h) 125. Zhang W et al. 2013 A novel core-shell
98. Rijal G, Li W. 2016 3D scaffolds in breast cancer 112. Du Y, Lo E, Ali S, Khademhosseini A. 2008 microcapsule for encapsulation and 3d culture of
research. Biomaterials 81, 135– 156. (doi:10.1016/j. Directed assembly of cell-laden microgels for embryonic stem cells. J. Mater. Chem. B Mater.
biomaterials.2015.12.016) fabrication of 3D tissue constructs. Proc. Natl Acad. Biol. Med. 2013, 1002–1009. (doi:10.1039/
99. Liu Z, Vunjak-Novakovic G. 2016 Modeling tumor Sci. USA 105, 9522–9527. (doi:10.1073/pnas. C2TB00058J)
microenvironments using custom-designed 0801866105) 126. Sakai S, Ito S, Inagaki H, Hirose K, Matsuyama T,
biomaterial scaffolds. Curr. Opin. Chem. Eng. 11, 113. McGuigan AP, Bruzewicz DA, Glavan A, Butte MJ, Taya M, Kawakami K. 2011 Cell-enclosing gelatin-
94 –105. (doi:10.1016/j.coche.2016.01.012) Whitesides GM. 2008 Cell encapsulation in sub-mm based microcapsule production for tissue
100. Kasuya J, Sudo R, Tamogami R, Masuda G, Mitaka T, sized gel modules using replica molding. PLoS ONE engineering using a microfluidic flow-focusing
Ikeda M, Tanishita K. 2012 Reconstruction of 3D 3, e2258. (doi:10.1371/journal.pone.0002258) system. Biomicrofluidics 5, 13402. (doi:10.1063/1.
stacked hepatocyte tissues using degradable, 114. Bruzewicz DA, McGuigan AP, Whitesides GM. 2008 3516657)
microporous poly(d,l-lactide-co-glycolide) Fabrication of a modular tissue construct in a 127. Yoon S, Kim JA, Lee SH, Kim M, Park TH. 2013
membranes. Biomaterials 33, 2693– 2700. (doi:10. microfluidic chip. Lab. Chip. 8, 663–671. (doi:10. Droplet-based microfluidic system to form and
1016/j.biomaterials.2011.12.039) 1039/b719806j) separate multicellular spheroids using magnetic
101. Pathi SP, Kowalczewski C, Tadipatri R, Fischbach C. 115. Matsunaga YT, Morimoto Y, Takeuchi S. 2011 nanoparticles. Lab. Chip. 13, 1522– 1528. (doi:10.
2010 A novel 3-D mineralized tumor model to study Molding cell beads for rapid construction of 1039/c3lc41322e)
128. Tsuda Y, Morimoto Y, Takeuchi S. 2010 130. Morimoto Y, Tan WH, Takeuchi S. 2009 Three- 132. Cui X, Liu Y, Hartanto Y, Bi J, Dai S, Zhang H. 2016 15
Monodisperse cell-encapsulating peptide microgel dimensional axisymmetric flow-focusing device Multicellular spheroids formation and recovery in

rsif.royalsocietypublishing.org
beads for 3D cell culture. Langmuir 26, 2645 –2649. using stereolithography. Biomed. Microdevices 11, microfluidics-generated thermoresponsive microgel
(doi:10.1021/la902827y) 369 –377. (doi:10.1007/s10544-008-9243-y) droplets. Colloid Interface Sci. Commun. 14, 4–7.
129. Morimoto Y, Tan WH, Tsuda Y, Takeuchi S. 131. Um E, Lee D-S, Pyo H-B, Park J-K. 2008 Continuous (doi:10.1016/j.colcom.2016.09.001)
2009 Monodisperse semi-permeable generation of hydrogel beads and encapsulation of 133. Chan HF, Zhang Y, Ho Y-P, Chiu Y-L, Jung Y, Leong
microcapsules for continuous observation of biological materials using a microfluidic droplet- KW. 2013 Rapid formation of multicellular spheroids
cells. Lab. Chip. 9, 2217– 2223. (doi:10.1039/ merging channel. Microfluid. Nanofluid. 5, in double-emulsion droplets with controllable
b900035f ) 541 –549. (doi:10.1007/s10404-008-0268-6) microenvironment. Sci. Rep. 3, 3462.

J. R. Soc. Interface 14: 20160877

You might also like