ArticlePDF Available

Three-Dimensional Cell Cultures in Drug Discovery and Development

Authors:

Abstract and Figures

The past decades have witnessed significant efforts toward the development of three-dimensional (3D) cell cultures as systems that better mimic in vivo physiology. Today, 3D cell cultures are emerging, not only as a new tool in early drug discovery but also as potential therapeutics to treat disease. In this review, we assess leading 3D cell culture technologies and their impact on drug discovery, including spheroids, organoids, scaffolds, hydrogels, organs-on-chips, and 3D bioprinting. We also discuss the implementation of these technologies in compound identification, screening, and development, ranging from disease modeling to assessment of efficacy and safety profiles.
Content may be subject to copyright.
https://doi.org/10.1177/2472555217696795
SLAS Discovery
1 –17
© 2017 Society for Laboratory
Automation and Screening
DOI: 10.1177/2472555217696795
journals.sagepub.com/home/jbx
Review
Introduction
Cell-based assays have been widely used in drug discovery
for several decades. Historically, two-dimensional (2D)
monolayer cells cultured on a variety of planar substrates
were the only practical option for cell-based screening and
have proven to be a convenient and effective means to dis-
cover drug candidate molecules. Nowadays, 2D cell models
can be used to effectively predict in vivo drug responses for
many targets and pathways and are still very useful in drug
discovery. However, it is evident that these 2D cultures suf-
fer disadvantages associated with the loss of tissue-specific
architecture, mechanical and biochemical cues, and cell-to-
cell and cell-to-matrix interactions,1,2 thus making them rela-
tively poor models to predict drug responses for certain
diseases such as cancer. For instance, compared with 2D cul-
ture, colon cancer HCT-116 cells in 3D culture have been
found to be more resistant to certain anticancer drugs such as
melphalan, fluorouracil, oxaliplatin, and irinotecan3; such che-
moresistance has been observed in vivo as well.4
The past decade has seen the accelerating implementa-
tion of 3D cell cultures in early drug discovery, principally
fueled by the need to continuously improve the productivity
of pharmaceutical research and development (R&D).5–7 The
use of 3D cell cultures, together with better cell models
such as stem cells and primary cells, would allow greater
predictability of efficacy and toxicity in humans before
drugs move into clinical trials,8,9 which, in turn, would
lower the attrition rate of new molecular medicines under
development. The 3D cell culture and co-culture models are
advantageous in that they not only enable drug safety and
efficacy assessment in a more in vivo–like context than tra-
ditional 2D cell cultures but also eliminate the species dif-
ferences (vs. animal models) that often impede interpretation
of the preclinical outcomes by allowing drug testing directly
in human systems.
In this review, we examine the new opportunities for the
application of 3D cell culture technologies in early drug dis-
covery, such as disease modeling, target identification and
validation, screening, and drug efficacy and safety assess-
ment. We also discuss emerging opportunities of 3D cell
cultures in drug development. Future directions and techni-
cal challenges for 3D cells-based drug discovery and devel-
opment are also discussed.
3D Cell Culture Technologies
Recent advances in cell biology, microfabrication tech-
niques, and tissue engineering have enabled the develop-
ment of a wide range of 3D cell culture technologies. These
include multicellular spheroids, organoids, scaffolds,
696795JBXXXX10.1177/2472555217696795SLAS DiscoveryFang and Eglen
research-article2017
1Biochemical Technologies, Corning Research and Development
Corporation, Corning Incorporated, Corning, NY, USA
2Corning Life Sciences, Corning Incorporated, Tewksbury, MA, USA
Received Sep 19, 2016, and in revised form Jan 28, 2017. Accepted for
publication Jan 30, 2017.
Corresponding Author:
Richard M. Eglen, Corning Life Sciences, Corning Incorporated, 836
North St., Building 300, Suite 3401, Tewksbury, MA 01876, USA.
Email: eglenrm@corning.com
Three-Dimensional Cell Cultures in Drug
Discovery and Development
Ye Fang1 and Richard M. Eglen2
Abstract
The past decades have witnessed significant efforts toward the development of three-dimensional (3D) cell cultures as
systems that better mimic in vivo physiology. Today, 3D cell cultures are emerging, not only as a new tool in early drug
discovery but also as potential therapeutics to treat disease. In this review, we assess leading 3D cell culture technologies
and their impact on drug discovery, including spheroids, organoids, scaffolds, hydrogels, organs-on-chips, and 3D bioprinting.
We also discuss the implementation of these technologies in compound identification, screening, and development, ranging
from disease modeling to assessment of efficacy and safety profiles.
Keywords
3D cell culture, 3D bioprinting, disease models, efficacy, organoids, organs-on-chips, safety, screening, multicellular
spheroid, toxicity
2 SLAS Discovery
hydrogels, organs-on-chips, and 3D bioprinting, each with
its own advantages and disadvantages (see Table 1 for a
summary). These 3D cultures, although different in princi-
ple and protocols, are used to restore the morphological,
functional, and microenvironmental features of human tis-
sues and organs. This section briefly describes the key fea-
tures of these technologies.
Spheroids
Multicellular spheroid cultures were initially developed by
Sutherland and coworkers in 1970 to recapitulate the func-
tional phenotype of human tumor cells and their responses
to radiotherapy.10,11 Since then, spheroid cultures have been
applied to many other types of cells, including stem cells,
hepatocytes, and neuronal cells (Table 1). Furthermore,
tumor spheroid monocultures or co-cultures with immune
or endothelial cells have been adapted to experimental can-
cer research and recently to oncology drug screening (see
below). The spheroid model compensates for many of the
deficiencies seen in monolayer cultures. For instance,
spheroids can develop gradients of oxygen, nutrients,
metabolites, and soluble signals, thus creating heteroge-
neous cell populations (e.g., hypoxic vs. normoxic, quies-
cent vs. replicating cells). In addition, spheroids have a
well-defined geometry and optimal physiological cell-cell
and cell-extracellular matrix (ECM) interactions. However,
there are several practical challenges associated with spher-
oid culture, including the development and maintenance of
spheroids of uniform size, the formation of spheroids from
a small seed number of cells, the precise control of specific
ratios of different cell types in spheroid when co-culture,
and the lack of reliable, simple, standardized, and high-
throughput compatible assays for drug screening using
spheroids.
There are four different approaches to enable spheroid cul-
tures. The first approach is to use low-adhesion plates to pro-
mote the self-aggregation of cells into spheroids12 (Fig. 1a).
These plates not only have an ultralow attachment surface
coating to minimize cell adherence but also possess a well-
defined geometry (e.g., round, tapered, or v-shaped bottom) to
drive and position a single spheroid within each well. The key
advantage of this approach is to form, propagate, and assay the
spheroids within the same plate, thus enabling high-throughput
screening (HTS) or high-content screening (HCS).
The second approach is to use hanging drop plates (HDPs)
to promote the formation of multicellular spheroids13 (Fig.
1b). When cells in media are dispensed into the top of an HDP
well, cells are segregated into the discrete media droplet
formed below the aperture of the HDP well bottom opening,
eventually forming spheroids. Similar to the low-adhesion
plates, the HDP can also be used for spheroid co-culture,
wherein multiple cell types are added either at the time of ini-
tial dispensing or sequentially. However, a clear caveat of this
approach is that spheroids are required to transfer from the
HDP to a second plate for assays.
The third approach is to use a bioreactor (e.g., spinner flask
or microgravity bioreactor) to drive cells to self-aggregate into
Table 1. Advantages and Disadvantages of Different 3D Cell Culture Techniques.
Technique Advantages Disadvantages
SpheroidsaEasy-to-use protocol
Scalable to different plate formats
Compliant with high-throughput screening (HTS)/high-content screening (HCS)
Co-culture ability
High reproducibility
Simplified architecture
Organoids Patient specific
In vivo–like complexity
In vivo–like architecture
Can be variable
Less amenable to HTS/HCS
Hard to reach in vivo maturity
Complication in assay
Lack vasculature
May lack key cell types
Scaffolds/hydrogels Applicable to microplates
Amenable to HTS/HCS
High reproducibility
Co-culture ability
Simplified architecture
Can be variable across lots
Organs-on-chips In vivo–like architecture
In vivo–like microenvironment, chemical, physical gradients
Lack vasculature
Difficult to be adapted to HTS
3D bioprinting Custom-made architecture
Chemical, physical gradients
High-throughput production
Co-culture ability
Lack vasculature
Challenges with cells/materials
Difficult to be adapted to HTS
Issues with tissue maturation
aDiscussion is limited to low-adhesion plates.
Fang and Eglen 3
spheroids under dynamic culture condition14 (Fig. 1c). This
approach permits large-scale production of spheroids.
However, this approach has disadvantages associated with flu-
idic flow-induced shear stress, as well as nonuniformity in size
of spheroids produced.
The fourth approach is to use micro-/nano-patterned sur-
faces as the scaffolds to control cell adhesion and migration,
thus enabling spheroid cultures15 (Fig. 1d). This approach
offers a wide range of nanoscale scaffolds imprinted onto a
flat substrate for the selection of appropriate patterns and
adhesive properties for a variety of cell types. Similar to
low-adhesion plates, these micropatterned plates have little
well-to-well and plate-to-plate variation, which make them
compliant with HTS. However, one caveat is that bubbles
may easily form during the culture, and pipetting often
damages the micropatterned surfaces.
Organoids
Organoids, also termed organ buds, represent a rapidly expand-
ing family of dish-based, 3D developing tissues that show real-
istic microanatomy.16–18 An organoid is “a collection of
organ-specific cell types that develops from stem cells or organ
progenitors and self-organizes through cell sorting and spa-
tially restricted lineage commitment in a manner similar to in
vivo.16 Organoids are classified into tissue and stem cell
organoids, depending on how the organ buds are formed.19
Tissue organoids refer to stromal cell–free (or mesenchyme-
free) culture and mostly apply to epithelial cells because of
their intrinsic ability to self-organize into tissue-like structures.
Stem cell organoids are generated from either embryonic stem
cells (ECSs) or induced pluripotent stem cells (iPSCs) or pri-
mary stem cells such as neonatal tissue stem cells or tissue-
resident adult stem cells. To date, several in vitro organoids
have been established to resemble various tissues, including
functional organoids for thyroid,20 pancreas,21 liver,22,23
stomach,24,25 intestine,26 vascularized cardiac patch,27 cerebral
cortex,28 thymus,29 kidney,30,31 lung,32 and retina.33 Table 2
summarizes key features (e.g., cell types culture techniques
used, and organotypic features) of these organoids.
Numerous different approaches have been used to obtain
organoids (see Table 2 for specifics).34 The first approach is
to directly culture cells as a monolayer on a bed of feeder
cells or an ECM-coated surface, so the organoids are formed
after the cells differentiate. The second approach is to use a
mechanically supported culture to allow the further differ-
entiation of primary tissues. For example, human keratino-
cytes can further differentiate and self-assemble into a fully
stratified tissue when the supported culture is in contact
with an air-liquid interface over a period of weeks.35 The
third approach is to generate embryoid bodies on the low-
adhesion plates or through hanging drop culture, similar to
spheroid cultures. The fourth approach is to use serum-free
floating culture of embryoid body-like aggregates with
quick reaggregation in low-adhesion plates.
Organoids mimic some, but not all, of the structure and
function of real organs.16 First, all organoids lack vascula-
ture, which is essential to nutrient and waste transport.
Second, some organoids may lack key cell types found in
vivo. Third, some organoids replicate only the early stages
of organ development. For example, retinal organoids do
not have the outer segments, and photoreceptors fail to fully
mature to become light sensitive, whereas the cerebral
organoids fail to fully develop later features, such as corti-
cal plate layers.16 Technical challenges still remain to pro-
duce organoids with in vivo–like complexity, increasing
maturity, and screening-compatible reproducibility.
Scaffolds and Hydrogels
Scaffolds refer to synthetic 3D structures made of a large
variety of materials with different porosities, permeability,
Figure 1. Four different techniques used for spheroid cultures. (a) A well of low-adhesion plates that have a round bottom with
an ultralow cell attachment coating.12 (b) A droplet of hanging drop plate where cells are partitioned and self-organized into a
spheroid.13 (c) Suspension culture in bioreactor where cells become self-aggregated into spheroids.14 (d) A representative pillar of
micropatterned plates where the cells are enriched on the top of the pillar to form a spheroid.15
4 SLAS Discovery
surface chemistries, and mechanical characteristics designed
to mimic the microenvironment of specific tissues. Scaffolds
can be classified into biological and polymeric scaffolds.
Biological scaffolds mostly use naturally derived ECM
such as Matrigel and collagen to promote appropriate cell
attachment and reorganization into 3D structures. Compared
to synthetic scaffolds, Matrigel can provide a more physio-
logically relevant microenvironment of soluble growth fac-
tors, hormones, and other molecules with which cells
interact in an in vivo environment.36 Matrigel has been
widely used as the gold standard scaffold material to pro-
vide 3D cell cultures for a wide range of cell types. However,
the disadvantages associated with Matrigel are commonly
occurring lot-to-lot variability during manufacturing and
complexity in composition, which are often ill-defined,
making it difficult to determine exactly which signals are
promoting cell function. Other natural gels such as fibrin,
hyaluronic acid, chitosan, alginate, or silk fibrils have also
been used for 3D cell culture; however, these natural gels
have less versatility to promote 3D culture than Matrigel.
Table 2. Organoids and Their Origin, Culture Techniques, and Applications.
Organoid Origin Culture Technique Endpoints Ref.
Thyroid mESCs EB differentiation in hanging
drops
Functional thyroid organoid 20
Pancreas Mouse embryo pancreas
progenitor
Matrigel embedding Epithelial derivatives including
endocrine cells
21
Liver mLGR5+ SC Matrigel embedding Bile ducts and hepatocytes to
model alpha-1 antitrypsin
deficiency and Alagille
syndrome
22
Liver hPSCs Co-culture with HUVECs and
hMSCs on Matrigel after
monolayer differentiation
toward endoderm
Liver bud derivative 23
Stomach Adult SC/gastric glands
(m/h)
Matrigel embedding Adult SC + all stomach epithelial
derivatives, excluding parietal
cells, to model Helicobacter
pylori infection/gastric cancer
24, 25
Intestine hESCs/PSCs Spheroids embedded Matrigel
after monolayer
differentiation toward hindgut
Intestinal bud, epithelial and
mesenchymal derivatives
26
Vascularized cardiac patch hESCs High FCS Contractile muscle 27
Cerebral cortex m/hESCs EBs generated in low-adhesion
U-shaped plates
Embedded in Matrigel and
cultured in spinner flask
Cerebral cortex to model
microcephaly
28
Thymus Fibroblasts Reprograming induced by
FOXN1
All types of thymic epithelial
cells on transplantation
29
Kidney hESCs/PSCs Subculture in air-liquid
interface after differentiation
and dissociation
Nephrons associated with
a collecting duct network
surrounded by renal
interstitium and endothelial
cells
30
Kidney hPSCs Sandwiched between two layers
of Matrigel, differentiation
with GSK3β inhibitor
Proximal tubules, podocytes,
and endothelium
31
Lung mAdult SCs Matrigel co-culture with lung
endothelial cells
Epithelial derivatives +
mesenchymal derivatives
32
Retina hESCs SFEBq in low-adhesion
V-shaped plates with
Matrigel embedding day 2,
transfer to Petri dish day 12
Epithelial + retinal derivatives 33
EB, embryonic body; ESCs, embryonic stem cells; FCS, fetal calf serum; FOXN1, transcription factor forkhead box N1; HUVECs, human umbilical vein
endothelial cells; LGR5, leucine-rich repeat containing G protein–coupled receptor 5; m/h, mouse or human; MSCs, human mesenchymal stem cells;
PSCs, induced pluripotent stem cells; SCs, stem cells; SFEBq, serum-free floating culture of EB-like aggregates with quick reaggregation.
Fang and Eglen 5
Polymeric scaffolds use synthetic hydrogels or other bio-
compatible polymeric materials to generate the physical
supports for 3D cultures.37,38 The hydrogels used for 3D
culture include poly(ethylene glycol) (PEG), poly(vinyl
alcohol), and poly(2-hydroxy ethyl methacrylate).38
Furthermore, hydrogels can be made to be hydrolytically or
enzymatically biodegradable by incorporating poly(lactic
acid) units39 or enzyme cleavable peptide sequences40 into
the polymer network backbone. The biodegradability is
critical to applications in which cell utilization is a must,
such as tissue engineering and regenerative medicine.
Synthetic scaffolds have several clear advantages over
Matrigel or other natural gels for 3D cultures. First, the use
of synthetic materials can minimize the relatively poor
reproducibility of biological ECMs between batches and
the resulting lack of consistency between cultures, as they
are often simply processed and manufactured. Second,
these scaffolds allow for fine tuning of biochemical and
mechanical properties, so it is possible to optimize both
mechanical and chemical cues for 3D cell cultures. Third,
these hydrogels possess high water content, enabling trans-
port of oxygen, nutrients, waste, and soluble factors, all of
which are important to cell functions.41 However, these
hydrogels do not contain the endogenous factors but act
mainly as a template to regulate cell behavior. In addition,
these hydrogels pose challenges related to oxygen availabil-
ity, heterogeneities present in the synthetic cellular micro-
environment, and uneven distribution of soluble growth
factors within the matrix and complication in imaging and
cell analysis.38
The scaffold characteristics, along with the material
properties, can regulate cell adhesion, proliferation, activa-
tion, and differentiation.42,43 For instance, naive mesenchy-
mal stem cells (MSCs) were shown to specify lineage and
commit to phenotypes with extreme sensitivity to substrate
mechanical stiffness.44 MSCs were neurogenic on soft
matrices but myogenic on stiffer matrices that mimic mus-
cle and osteogenic on comparatively rigid matrices that
mimic collagenous bone. Upon treatment with soluble fac-
tors, MSCs were found to differentiate into the lineage
specified by matrix elasticity.
Scaffolds can be made using a variety of techniques,
such as 3D printing,45 particulate leaching,46 or electrospin-
ning.47 Alternative approaches include gas foaming, fiber
meshes/fiber bonding, phase separation, melt molding,
emulsion freeze drying, solution casting, or freeze drying
(reviewed in ref. 48). The types of scaffolds obtained were
dependent on the fabrication techniques. In general, partic-
ulate leaching or solvent casting can be used to produce
porous scaffolds, whereas electrospinning is useful for fab-
ricating fibrous scaffolds, and 3D printing can be used to
produce scaffolds with defined shapes and geometries. All
of these types of scaffolds have been realized for 3D cul-
ture. For instance, to overcome the progressive loss of
functionality of MSC expansion in 2D monolayer culture,
freshly isolated bone marrow nucleated cells were directly
cultured within 3D porous hydroxyapatite ceramic scaf-
folds in a perfusion-based bioreactor system.49 The stromal
tissues obtained were enzymatically treated to yield CD45-
MSCs, which gave rise to a 4.3-fold higher clonogenicity
and the superior differentiation capacity toward all typical
mesenchymal lineages, compared with the 2D expansion
culture. Of note, cells grown on fibrous scaffolds are often
not considered to truly represent 3D culture, as cells typi-
cally adhere and elongate along the fibers.50 In addition,
porous scaffolds have issues associated with their limited
diffusion properties, which make it difficult to fabricate
more complex tissues such as heart and liver.
Organs-on-Chips
An organ-on-a-chip refers to an artificial, miniature model
of a human organ on a microfluidic cell culture chip. The
chip is made with great precision using microfabrication
techniques such as soft lithography, photolithography, and
contact printing.51 The chip usually consists of a series of
well-defined structures, patterns, or scaffolds. Therefore,
the position, shape, function, and chemical and physical
microenvironments of the cells in culture can be controlled
with high spatiotemporal precision using microfluidics.52
Organs-on-chips are designed to reconstitute the structural,
microenvironmental, and functional complexity of living
human organs. However, most organs-on-chips are often
made to capture only the critical features of an organ type or
a disease model due to practical reason, so researchers can
reproduce clinically relevant disease phenotypes and phar-
macological responses.53,54 To date, a wide range of organs-
on-chips have been reported, including skin,55 lung,56–58
vasculature,59 heart,60,61 muscle,62 liver,63–65 intestine,66 and
several others (see below for specific applications of some
of these organ systems).
Organs-on-chips have been adapted to microplate for-
mats, the de facto footprint used in drug discovery. For
instance, a liver-on-a-chip that uses a bioreactor to foster
maintenance of 3D tissue cultures under constant perfusion
was developed in a multiwell plate format67 and used for
drug metabolism profiling and pharmacokinetic evalua-
tion.68 However, most organs-on-chips lack vasculature and
also are difficult to adapt to HTS.
Three-Dimensional Bioprinting
Three-dimensional bioprinting refers to the printing of
cells, biocompatible materials, and supporting components
into complex 3D living tissues with the desired cell/organ-
oid architecture, topology, and functionality using additive
manufacturing.69 Three-dimensional bioprinting usually
involves layer-by-layer positioning of biological materials,
6 SLAS Discovery
biochemicals, and living cells. There are three approaches
used for bioprinting. The first one is biomimicry, which
employs biologically inspired engineering to replicate the
cellular and extracellular components of a tissue or organ
(e.g., human ears).70 The second approach is autonomous
self-assembly, which relies on the cells as the primary driver
of histogenesis to produce the desired biological microar-
chitecture and functional tissues.71 The third approach is to
fabricate and assemble mini-tissue building blocks, such as
a kidney nephron, into the larger construct by rational
design, self-assembly, or a combination of both.72,73
Three-dimensional bioprinting has been used to generate
functional tissues, such as multilayered skin, bone, vascular
grafts, tracheal splints, heart tissue, and cartilaginous struc-
tures, for transplantation applications.74 Furthermore, 3D bio-
printing has been used not only to create scaffolds for 3D cell
cultures but also to directly produce 3D-bioprinted tissue mod-
els for drug screening and profiling.75 Bioprinting has several
advantages, such as custom-made microarchitecture, high-
throughput capability, and co-culture ability. However, com-
pared with other 3D cell cultures, 3D bioprinting faces many
additional challenges associated with cell and material require-
ments as well as tissue maturation and functionality.69
3D Cell Cultures in Drug Discovery
Drug discovery is a long, complex process with growing dif-
ficulty. Three-dimensional cell cultures have been penetrating
into the early drug discovery process, starting from disease
modeling to target identification and validation, screening,
lead selection, efficacy, and safety assessment (Fig. 2). This
section discusses how to best implement different 3D cell cul-
ture technologies into different stages of drug discovery
process.
Disease Modeling
Drug discovery often starts with a disease or a clinical con-
dition without suitable medical products available.76 As
growing efforts have been directed toward unmet therapeu-
tic needs in recent years,77 disease modeling has become
increasingly important to the success of drug discovery pro-
grams. As they promise to bridge the gap between 2D cul-
ture and in vivo, a range of 3D cell cultures have been
applied to understand the mechanisms of different diseases.
In particular, 3D models have gained popularity in elucidat-
ing tumor biology, as standard 2D models are inadequate to
address questions regarding indolent disease, metastatic
colonization, dormancy, relapse, and the rapid evolution of
drug resistance.78
Three-dimensional cultures on ECM gels have provided
models to detect architecture transformation from preinva-
sive breast carcinoma to full malignancy induced by the
progressive loss of tissue architecture and aberrant signal-
ing79 or from nonmalignant breast epithelial cells to malig-
nant tumors induced by tuning stiffness of Matrigel/
Collagen I gels used in 3D culture.80 Recently, Drost et al.81
investigated the phenotypes of sequential cancer mutations
in cultured human intestinal stem cells by combining organ-
oid culture and CRISPR/Cas9 gene editing. Here, normal
human intestinal stem cells isolated from patients were
genetically edited using CRISPR/Cas-9 for the four most
commonly mutated colorectal cancer genes (APC, P53,
KRAS, and SMAD4), followed by culturing on Matrigel or
basal membrane extract–coated plates in medium contain-
ing the stem-cell-niche factors WNT, R-spondin, epidermal
growth factor, and noggin. Results showed that the epithe-
lial organoids obtained remained genetically and phenotyp-
ically stable for long periods of time, and xenotransplantation
of quadruple mutant organoids into mice resulted in tumors
with features of invasive carcinoma. Remarkably, the com-
bined loss of APC and P53 was found to be sufficient for the
appearance of extensive aneuploidy, a hallmark of tumor
progression.
Spheroid cultures have become useful for modeling the
tissue architecture, signaling, microenvironments, and inva-
sion and immune behaviors of cancer, as well as for study-
ing and expanding the cancer stem cells (CSCs).82 Human
Figure 2. How different three-
dimensional culture techniques
have been implemented in
different stages of drug discovery
and development processes.
Representative references for
each application are cited in
parentheses in the graph.
Fang and Eglen 7
cancer is known to harbor several heterogeneous subpopu-
lations of CSCs that play distinct roles in tumor initiation,
maintenance, and metastasis. For instance, in colon cancers,
there were three types of CSCs isolated from patients:
tumor-initiating cells that have limited or no self-renewal
capacity but are contributed to tumor formation only in pri-
mary mice, self-renewal CSCs that allow long-term tumor
growth, and rare delayed contributing CSCs that were
exclusively active in secondary or tertiary mice.83 Tumor
invasion and metastasis is a multistep cascade process. It
begins with local invasion of cancer cells through the ECM
and stromal cell layers, then intravasation into the lumina of
blood vessels. This is followed by transit through the lym-
phatic and hematogenous systems and arrest and extravasa-
tion out of the circulatory system, which leads to the
formation and growth of micrometastatic lesions into mac-
roscopic tumors at a distant site.84 Spheroids of cancer cell
lines have been used to investigate different aspects of the
cancer invasion process, including the invasion of cells in a
3D spheroid into the surrounding 3D ECM structure85,86 and
endothelial cell–tumor cell interactions.87 For instance, we
had developed a label-free, real-time, single-cell, and quan-
titative assay to monitor the invasion of cells in a spheroid
through a 3D Matrigel (Fig. 3). We found that epidermal
growth factor accelerates the invasion of the colon cancer
cell line HT-29, whereas vandetanib dose-dependently
inhibits the invasion.88 Vandetanib is a multitarget kinase
inhibitor that has been clinically approved for the treatment
of late-stage (metastatic) medullary thyroid cancer in adult
patients who are ineligible for surgery and also has potential
to treat non–small-cell lung cancer. Although the results
obtained using the label-free assay are largely expected as
vandetanib is known to inhibit vascular endothelial growth
factor receptor, this assay enables real-time quantification
of its effect on cancer invasion through the Matrigel, a capa-
bility that is otherwise difficult to obtain using conventional
endpoint assays. We further found that PTEN knockout
increased the invasion rate of HCT116 cells in spheroid
through 3D Matrigel, and PI3K inhibitors LY294002 and
wortmannin drastically reduced the invasiveness of the
Figure 3. A label-free, single-cell, real-time assay to measure the invasion of cells in a single spheroid through a three-dimensional
extracellular matrix (Matrigel). (a) Principle of the assay, which consists of four critical steps: coating the biosensor surface with
Matrigel; adding medium to the well; transferring a spheroid from an ultralow attachment, round-bottomed microplate and placing
it onto the top Matrigel surface; and monitoring the invasion of cells through the matrix and adhesion on the sensor surface in
real time. (b–d) The time series dynamic mass redistribution (DMR) images before and after a single spheroid was placed onto the
biosensor surface coated with 10 µL 0.1 mg/mL Matrigel: 0 min (b), 1 h (c), and 24 h (d). Spatial scale bar: 500 µm. Intensity scale bar:
–500 pm to 2000 pm. (e) A DMR image taken 24 h after a spheroid was placed on the top Matrigel surface. Scale bar: 500 µm. (f)
Representative pixelated real-time DMR signals for the black line indicated in (e). (g) The adhesion events versus cell types. (h) The
adhesion time to reach 200 pm under different conditions. For (e–h), coating was 0.2 mg/mL Matrigel. Data represent mean ± SD for
g (n = 3). ***p < 0.001. This figure is adapted from ref. 89 with permission.
8 SLAS Discovery
cells.89 This label-free imaging technique has revealed that
besides the accelerated invasion kinetics, PTEN knockout
expedites cell dissociation from the spheroidal structure and
adhesion onto the surfaces. This study also indicates that the
mechanisms governing cell invasion are sensitive to ECM
matrix density, and the invasion inhibitory sensitivity of
PI3K inhibitors is also sensitive to the PTEN expression
level.
Organoid cultures have also been applied to model can-
cer, besides a great number of other diseases including
developmental disorders, infectious diseases, and neuronal
degeneration.16 For example, several different intestinal
organoids were obtained and used for modeling a range of
diseases. Specifically, the human intestinal organoids
derived from the ESC line WA09 were used to examine gas-
trointestinal infection with rotavirus.90 The human intestinal
organoids generated using intact crypts from human intes-
tines were used to examine Cryptosporidium parvum infec-
tion.91 The organoids obtained by culturing CD44+CD24+
cells enriched for colorectal CSCs in the HT29 and SW1222
cell lines were used to study colon CSC biology.92 The
intestinal organoids obtained using murine primary intesti-
nal cells were used to study genetically reconstituted tumor-
igenesis (e.g., by knockdown adenomatous polyposis coli
[APC]),93 whereas the intestinal organoids cultured from
patient biopsies were used to study genetic disorders.94,95
Many genetic disorders that have been difficult or impos-
sible to model in animals can be modeled by using organoid
cultures of patient iPSCs or, alternatively, through the
introduction of patient mutations into human PSCs using
genome-editing technologies, such as CRISPR/Cas9. For
instance, the CRISPR-Cas9 genome-editing system was
used recently to introduce multiple recurrent mutations in
colon cancer patients into organoids derived from normal
human intestinal epithelium.96
Organs-on-chips are also useful for cancer modeling.
For instance, cultured human skin tissue has been success-
fully used as a surrogate for modeling melanoma cancer
growth.55 Here, when human melanoma cell lines were
incorporated, the cultured skin tissue recapitulated natural
features of melanocyte homeostasis and melanoma progres-
sion in human skin. They displayed the same characteristics
reflecting the original tumor stage (vertical and radial
growth phases and metastatic melanoma cells) in vivo.
Organs-on-chips have also been used to model other dis-
eases. For instance, a lung-on-a-chip was developed to
mimic breathing by stretching and compressing an artificial
alveolar-capillary barrier using a cyclic vacuum machine.
This was used to model pathogen infection and inflamma-
tory responses to air pollutants56 or the development and
progression of pulmonary edema induced by the toxicity of
interleukin-2.57 Recently, the airway-on-a-chip device lined
by living human bronchiolar epithelium from normal or
chronic obstructive pulmonary disease (COPD) patients
was connected to an instrument that “breathes” whole ciga-
rette smoke in and out of the chips to study smoke-induced
pathophysiology in vitro.58 This enables the detection of
smoke-induced ciliary micropathologies, COPD-specific
molecular signatures, and epithelial responses to smoke
generated by electronic cigarettes.
Target Identification and Validation
Target identification and validation is often the rate-limiting
step in preclinical drug discovery.97 Three-dimensional cul-
tures have the potential to discover novel mechanisms and
targets and to accelerate target identification and validation,
given that the gene expression patterns found in 3D models
are one step closer to in vivo, compared to 2D monolayer
models.98 For instance, gene expression analysis of meso-
thelioma cell lines cultured in spheroids had revealed the
underlying causes of chemoresistance in malignant pleural
mesothelioma.99 Here, the spheroids were found to acquire
increased chemoresistance compared with 2D monolayers.
A total of 209 genes were differentially expressed in com-
mon by the three mesothelioma cell lines in spheroids,
among which argininosuccinate synthase 1 (ASS1) was the
only consistently up-regulated gene in both 3D spheroids
and human tumors. siRNA knockdown of ASS1 signifi-
cantly sensitized mesothelioma spheroids to the proapop-
totic effects of bortezomib or cisplatin plus pemetrexed.
These results suggest that ASS1 may be a druggable target
to undermine mesothelioma multicellular resistance.
In another recent study, a microfluidic vasculature chip
was developed to model intravascular steps in metastasis.59
Here, the chip consisted of an upper intravascular compart-
ment and lower stromal chambers, separated by a semipo-
rous membrane lined with human microvascular endothelial
cells. Upon stimulation of microvascular endothelium from
the basal side, CXCL12 acted through the CXCR4 receptor
on endothelium to promote adhesion of circulating breast
cancer cells. This suggests that targeting CXCL12-CXCR4
signaling in endothelium may limit metastases in breast and
other cancers.
Screening for Hit Identification
Screening using cell-based assays has frequently been the
starting point for identifying hit compounds in the early
stage of drug discovery. In the past three decades or so,
target-based HTS has been dominating in the hit identifica-
tion process, given that HTS-compatible cellular assays
have simplicity, relatively low cost, and high efficiency.
However, in recent years, there has been a renaissance in
phenotypic screening, driven by three factors. First, con-
tinuous improvement in the productivity of pharmaceutical
R&D calls for innovative strategies for drug discovery.
Second, although target-based screens are more effective
Fang and Eglen 9
for discovering follow-on drugs for which molecular mode
of action is known, phenotypic screens are more productive
for discovering first-in-class drugs.100 Third, advances in
detection technologies have made it feasible to perform
phenotypic screens with high throughput as well as more
biologically relevant information relative to conventional
molecular assays.101–104
Incorporating 3D cell cultures with HTS processes is
still in infancy but shows promise in directly identifying
clinically relevant compounds, enabling effective transla-
tional research. Unfortunately, not all 3D cell culture mod-
els are compatible with HTS or HCS in a routine and
cost-effective manner. Among all 3D models under devel-
opment, spheroids cultured in the low-adhesion plates have
started gaining popularity in oncology drug screening
because of their easy-to-use protocols, high-density micro-
plate formats (e.g., 384-well and 1536-well), and compati-
bility with automation and multimode detection systems.
For instance, using glucose-deprived multicellular tumor
spheroids of colon cancer cell lines with inner hypoxia that
were cultured in 384-well low-adhesion plates, Senkowski
et al.105 screened 1600 compounds with documented clini-
cal history to identify five compounds that selectively target
the hypoxic cell population. All five compounds inhibited
mitochondrial respiration, suggesting that cancer cells in
low-glucose concentrations depend on oxidative phosphor-
ylation, instead of solely glycolysis. The antiprotozoal drug
nitazoxanide was found to activate the AMPK pathway and
down-regulate c-Myc, mTOR, and Wnt signaling at clini-
cally relevant concentrations. Combining nitazoxanide with
the cytotoxic drug irinotecan showed anticancer activity in
vivo. Similar results were obtained from the HCS of 1120
compounds against spheroids of the human breast cancer
cell line T47D.106 At the 2016 SLAS annual conference, Dr.
Timothy Spicer and his colleagues at The Scripps Research
Institute presented results using Corning nonadherent 1536-
well spheroid plates to screen the entire Scripps Drug
Discovery Library of more than 650,000 compounds in less
than 2 wk (personal communication).
Three-dimensional co-cultures of a cancer cell with
another cell type (e.g., an immune or fibroblast cell line)
have also been developed in high-throughput formats. For
instance, using a multilayered organotypic culture contain-
ing primary human fibroblasts, mesothelial cells, and ECM,
Kenny et al.107 performed a screen of 2420 pharmacologi-
cally active compounds. This organotypic culture was used
to reproduce the human ovarian cancer metastatic microen-
vironment. Subsequent validation in secondary in vitro and
in vivo assays confirmed two active compounds, β-escin
and tomatine, that prevented ovarian cancer adhesion, inva-
sion, and metastasis, leading to the improved survival in
mouse models. This study shows the power of complex 3D
models to improve the disease relevance of assays used for
drug screening.
Efficacy Profiling for Lead Identification
Following hit identification is lead identification. Once
identified in a screen, hits are first confirmed based on
dose-response curves using the same assay for screening
and orthogonal testing with different assay(s). Once con-
firmed, hits are further evaluated for synthetic tractability,
freedom to operate, drug-likeness, and possible toxicity,
metabolism, and stability-related risks. Medicinal chemis-
try optimization is the next step to generating lead candi-
date compounds with improved potency, reduced off-target
activities, and desired physicochemical and metabolic
properties. Critical to the entire process of lead identifica-
tion is to have cost-effective in vitro models that can more
reliably predict the efficacy, toxicity, and pharmacokinet-
ics of drug compounds in humans. Three-dimensional cell
culture models have a potential to play an important role
in lead identification and to reduce the use of animal test-
ing for preclinical studies.
Lacking in vivo efficacy is one of the key reasons why
some late-stage clinical trials fail.108 Three-dimensional cell
culture models have been shown to in some cases more accu-
rately evaluate drug efficacy than 2D models and may even
enable personalized approaches to identify the mechanisms
underlying disease and to screen and select the best drug(s)
for the patients.109–111 For instance, patient-derived spheroids
have been developed as a predictive test to identify the most
effective therapy for 120 patients with HER2-negative breast
cancer of all stages.112 Results showed that the tissue spher-
oid model reflected current guideline treatment recommen-
dations for HER2-negative breast cancer. Tissue spheroid
showed greater responses to anthracycline/docetaxel for hor-
mone receptor–negative samples, a higher response to fluo-
rouracil and anthracycline in high-grade tumors, and a higher
treatment efficacy to anthracycline treatment combined with
fluorouracil for smaller tumor size and negative lymph node
status. Recently, Tong et al. applied spheroids of three ovar-
ian cancer cell lines to investigate the differential oncolytic
efficacy among three different viruses: myxoma, double-
deleted vaccinia, and Maraba virus.113 They found that the
low-density lipoprotein receptor expression in ovarian cancer
spheroids is reduced, which in turn affects the binding and
entry of Maraba virus into cells.
Compared with spheroids, organs-on-chips provide a
viable strategy to further increase the complexity and physi-
ological relevance for reliable assessment of drug efficacy.
For instance, Aref et al.114 developed an organ-on-a-chip
consisting of lung cancer spheroids in a 3D matrix gel adja-
cent to an endothelialized microchannel to recapitulate
epithelial-mesenchymal transition during cancer progression.
Results showed that for the A549 cell model, there are both
qualitative and quantitative differences in drug response
between 2D monolayer cells and 3D spheroids. For instance,
for the TGF-βR inhibitor A83-01, the differences in
10 SLAS Discovery
effective dose between 2D and 3D culture were more than
three orders of magnitude (5 nM vs. 2.5 μM).
Toxicity Profiling for Lead Selection
Drug-induced toxicities in liver, heart, kidney, and brain
currently account for more than 70% of drug attrition and
withdrawal from the market.115 Adverse drug reactions are
often due to off-target interactions or excessive binding of
the drug molecule to toxicity-prone cells. Three-dimensional
cell culture models are powerful in assessing drug-induced
toxicity.
Organ buds of brain, liver, heart, and kidney can be used to
assess drug toxicity.16 Recently, a brain organoid was produced
by combining human ESC-derived neural progenitor cells,
endothelial cells, MSCs, and microglia/macrophage precur-
sors on chemically defined polyethylene glycol hydrogels.116
Machine learning was used to build a predictive model from
changes in global gene expression when being exposed to 60
training compounds (34 toxic and 26 nontoxic chemicals). The
model was then used to correctly classify 9 of 10 additional
chemicals in a blinded trial. Human liver organoids obtained
using HepaRG cell line, a terminally differentiated hepatic cell
line derived from a human hepatic progenitor cell line, have
already been shown to produce human-specific metabolites.117
This is particularly useful because human liver often metabo-
lizes drugs in a manner distinct from animal liver. Of note,
these HepaRG 3D organotypic cultures are more sensitive to
acetaminophen- or rosiglitazone-induced toxicity but less sen-
sitive to troglitazone-induced toxicity than the 2D cultures.
Kidney organ buds from human iPSC cells were found to dif-
ferentially apoptose in response to cisplatin, a nephrotoxicant,
showing such organoids represent powerful models of the
human organ for drug-induced nephrotoxicity.30
Three-dimensional liver cell spheroid cultures are also
valuable for investigating drug-induced liver injury, func-
tion, and diseases. An organotypic culture of the human
hepatoma HepaRG cell line were obtained using hanging
drop culture and was able to detect the potent toxicity of
acetaminophen.117 Human primary hepatocyte spheroids
obtained using the low-adhesion plates were found to be
phenotypically stable and retained morphology, viability,
and hepatocyte-specific functions for at least 5 wk, enabling
chronic toxicity assessment of drug molecules.118 The
chronic toxicity of fialuridine was detected after repeated
dosing in this spheroid model; this type of toxicity was
impossible to detect using 2D models. However, the pri-
mary hepatocyte spheroids also retain the interindividual
variability, which may limit the ability of such models for
large-scale screening. To this regard, unlimitedly renew-
able, primary-like hepatocytes, such as HepatoCells,
HepRG, or iPS-derived cells, may be good alternatives for
screening.
Organs-on-chips and other 3D cell culture models were
also used to evaluate drug-induced toxicity.104 Heart-on-a-
chip devices were useful for assessing drug-induced cardio-
toxicity.60,61 The lung-on-chip model developed by Huh
et al.57 consisted of channels lined by closely apposed layers
of human pulmonary epithelial and endothelial cells that
experience air and fluid flow, enabling the detection of drug
toxicity-induced pulmonary edema observed in human can-
cer patients treated with interleukin-2 at similar doses and
over the same time frame.57 This study also found that both
angiopoietin-1 and GSK2193874 (a transient receptor
potential vanilloid 4 ion channel inhibitor) were effective at
preventing the drug toxicity-induced pulmonary edema. A
3D bioprinted, cell-based soft robotic device that was pow-
ered by the actuation of an engineered mammalian skeletal
muscle strip was recently used to sense, process signals, and
produce force.62 The muscle strip was made by printing
mouse skeletal muscle myoblast cell line C2C12 in the
presence of hydrogels and other biological components.
Skeletal muscle as a contractile power source is the primary
generator of actuation in animals. This device can be used
to assess drug-induced myopathy.
Pharmacokinetics and Pharmacodynamics
Profiling for Lead Selection
Inadequate pharmacokinetics and pharmacodynamics is
also a key factor in why drugs fail. Three-dimensional cell
culture models, in particular, liver spheroids, liver organ-
oids, and body-on-chips, are useful to investigate the phar-
macokinetic profiles of drug molecules. Liver spheroids
and organoids have been used to study the metabolism of
drug molecules.116 Several versions of liver-on-a-chip sys-
tems were used to measure rates of metabolic drug clear-
ance, which were compared with literature-reported
values.63–65 The gut-on-a-chip using the Caco-2 cell layer
on a porous support to separate two chambers was used to
reproduce characteristic absorptive properties and the bar-
rier function of the human intestine, enabling drug absorp-
tion studies.66 Integrating multiple organ types into one
chip, termed as body-on-a-chip, can be powerful for com-
prehending the pharmacokinetics and pharmacodynamics
of drug molecules.119,120 However, developing screening-
compatible body-on-a-chip remains a challenging task, in
particular when one considers the known allometric scaling
issue.121
3D Cultures in Cell Therapy and
Tissue Engineering
Cell therapy and tissue engineering have started entering
the market. They not only offer new hope for patients with
injuries, end-stage organ failure, or other clinical issues but
Fang and Eglen 11
also will eventually transform our lives. However, it is
becoming clear that realizing the full potential of cell ther-
apy and tissue engineering requires advances in cell culture
technologies to meet the demand in quantity, quality, and
process robustness for commercialization and clinical trials.
Three-dimensional cell cultures offer not only a solution for
cell scale-up production but also a new form of therapeutics
for treating many different diseases.
Stem Cell Spheroids for Regenerative Medicine
Stem cells are widely used as a cell source for regenerative
medicine and cell therapy applications. However, conven-
tional 2D culture techniques, in combination with the cur-
rent best practice, may be ineffective to expand stem cells
for clinical applications. This is reflected by the fact that 2D
cultures are inadequate to reproduce the in vivo microenvi-
ronment of stem cells.122 In addition, clinical observations
show that the beneficial effects of stem cell–based thera-
peutics seen in initial small-scale clinical studies are often
not validated by large, randomized clinical trials.123,124 In
fact, MSCs often decrease their replicative ability, colony-
forming efficiency, and differentiation capabilities over
time when culturing and passaging in 2D adherent mono-
layer.125,126 In contrast, MSCs cultured in spheroids display
a morphology that is significantly different from 2D cul-
ture.127 The MSCs are spherical inside and elongated out-
side the spheroid, with an overall reduction of cytoskeletal
molecules, ECM, and size (~75% reduction in individual
cell volume),128 indicating distinct differentiation prefer-
ences among different lineages.129 Furthermore, compared
with 2D culture, MSCs cultured in spheroids have different
gene expression patterns, with up-regulation of many genes
that are associated with hypoxia, angiogenesis, inflamma-
tion, stress response, and redox signaling.130
Spheroid cultures have been reported to improve the
efficacy of MSC-based therapeutics. Compared with 2D
cultures, MSC spheroid cultures were found to result in sev-
eral additional beneficial effects, such as enhanced anti-
inflammatory and tissue regenerative and reparative effects,
as well as better posttransplant survival of MSCs.130
Furthermore, compared with 2D cultured cells, spheroids of
human adipose–derived MSCs produced higher levels of
ECM proteins, exhibited stronger antiapoptotic and antioxi-
dative capacities, and increased the paracrine secretion of
cytokines.131 When injected into the kidney of model rats
with ischemia reperfusion-induced acute kidney injury,
these MSC spheroids were more effective in protecting the
kidney against apoptosis, reducing tissue damage, promot-
ing vascularization, and ameliorating renal function com-
pared with 2D cultured cells.
Spheroid cultures have been used to enrich patient-
specific stem cells for disease treatment. For instance,
Henry et al.132 applied spheroid culture to enrich adult lung
stem cells for use in treating idiopathic pulmonary fibrosis
in mice. Here, in a suspension culture, the outgrowth cells
from healthy lung tissue explants were self-aggregated into
spheroids, which recapitulated the stem cell niche and
acquired mature lung epithelial phenotypes. The mice that
received these spheroids showed decreases in inflammation
and fibrosis.
Spheroid cultures have also been used to scale up stem
cell products for use in clinical trials. For instance, the man-
ufacturing process of pancreatic endoderm cells (PEC-01)
involves dynamic suspension spheroid culture and differen-
tiation.133 The PEC-01 is derived from CyT49 human ESCs
and is the cellular component of the VC-01 combination
product from ViaCytes for treating type 1 diabetes. PEC-01
matures after transplantation and functions to regulate
blood glucose.
Organoids for Transplantation
Organoids could provide a source of autologous tissue for
transplantation, as organoid research advances rapidly. For
instance, renal organoids derived from pluripotent stem
cells were successfully transplanted under the renal cap-
sules of adult mice.134 Here, the organoid reconstituted the
3D structures of the kidney in vivo, including glomeruli
with podocytes and renal tubules with proximal and distal
regions and clear lumina. Furthermore, the glomeruli were
efficiently vascularized upon transplantation, which is a
promising step toward kidney replacement strategy.
Although early in development, organoid-based replace-
ment may find applications in other diseases, such as retinal
organoids obtained from human ESCs for treating certain
types of retinal degeneration and blindness,16 intestinal
organoids for replacement of damaged colon after injury or
following removal of diseased tissue,135 and gene-corrected
organoids for replacement of damaged organs with gene
defect(s).16 For instance, the intestinal organoids obtained
from Lgr5+ adult colonic stem cells have been transplanted
into superficially damaged mouse colon.136 Results showed
that the transplanted donor cells readily integrated into the
mouse colon, covering the area that lacked epithelium as a
result of the introduced damage in recipient mice. Long-
term (>6 months) engraftment with transplantation of
organoids derived from a single Lgr5+ colon stem cell was
observed after extensive in vitro expansion. This study
shows the feasibility of colon stem-cell therapy based on
the in vitro expansion of a single adult colonic stem cell.
3D Bioprinted Tissues/Organs for
Transplantation
Advances in tissue engineering, cell biology, and materials
sciences have made 3D bioprinting possible to create
functioning tissues or organ grafts with their natural
12 SLAS Discovery
microenvironments and architectures from autologous cells
for transplantation applications. Although printing an intact
organ still remains elusive, 3D-bioprinted bladders, tracheal
grafts, bone, and cartilage have proven to be functional after
development and implantation in animals or humans.74
These printed organs can be used as assist organs or viable
replacements. For instance, Atala et al.137 engineered a
human bladder by isolating autologous bladder urothelial
and muscle cells from the bladder biopsy, expanding the
cells in vitro and seeding them to a biodegradable bladder-
shaped scaffold made of collagen or a composite of colla-
gen and polyglycolic acid. About 7 weeks after the biopsy,
the autologous engineered bladder constructs were used for
reconstruction and implanted either with or without an
omental wrap. A clinical trial on seven patients in need of a
cystoplasty showed that the engineered bladder tissues, cre-
ated with autologous cells seeded on the collagen-polygly-
colic acid scaffolds and wrapped in omentum after
implantation, were safe and effective to use in patients. In
another example, a microfluidic device with double-coaxial
laminar flow was used to fabricate meter-long core-shell
hydrogel microfibers encapsulating ECM proteins and pri-
mary pancreatic islet cells.138 After transplanting through a
microcatheter into the subrenal capsular space of diabetic
mice, the microfibers containing the islet cells normalized
blood glucose concentrations for about 2 wk.
More recent efforts were focused on the development of
3D-bioprinted tissues, such as livers and kidneys, with inte-
grated vasculature.139 Integral vascular structures are criti-
cal to the survival of the transplanted organs or tissues.
Either autologous vascular conduits from deceased donor or
synthetic vascular grafts have been used for anastomosing
the new organ to the recipient when necessary; however,
both come with disadvantages. Printing using spheroids of
human umbilical vein smooth muscle cells and human skin
fibroblasts, along with agarose rods, has resulted in single-
and double-layered vascular tubes with small diameters.140
Furthermore, printing branched vascular structures using
human umbilical vein endothelial cells, 10T1/2 cells,
human fibroblasts, or human embryonic kidney cells is also
feasible.141,142
Challenges, Limitations, and Future
Perspectives of 3D Cell Cultures
Many challenges remain for the widespread adoption of 3D
cell culture technologies in the drug discovery process. In
fact, there are very limited 3D screens done with large com-
pound libraries, although a multitude of 3D assays, mostly
based on high-resolution fluorescence imaging techniques,
have been validated for HTS/HCS in recent years.143,144
First, many 3D models such as organoids exhibit signifi-
cantly more complex morphology and function than 2D
cultured cells, thus leading to challenges in systematic
assessment. Furthermore, current 3D cultures are diverse in
terms of complexity, size, morphology, 3D architecture, and
protocols for assaying. This leads to challenges in standardiza-
tion with respect to culture and assay protocols, phenotypes,
and output data for analysis. To this regard, the development of
high-density microtiter plate–based spheroid-forming plates
(e.g., 1536-well low-adhesion spheroid plates) represents
an attractive solution to streamline 3D spheroid-based drug
screening. The 1536-well spheroid-forming plates also
make HTS economically affordable.
Second, lacking the understanding of the relevance of a
3D phenotype measured to the in vivo drug effects sought
also possesses challenges for 3D screening, as typical 3D
assay techniques measure a wide range of cellular pheno-
typic parameters (e.g., spheroid size or morphology,
hypoxic core). As the mainstay in 3D assays, high-content
imaging techniques could measure many different pheno-
types. However, identifying a clinically relevant phenotype
that is measurable in 3D models is critical to streamline and
expedite the screening process. For instance, using spher-
oids of invasive human prostate cancer cell line PC3 cul-
tured in the Matrigel matrix, Booij et al.144 developed a
phenotypic imaging assay to measure more than 800 pheno-
typic parameters. Multiparametric analysis identified sev-
eral phenotypes that enable the discrimination of selective
inhibitors for c-Met, or epidermal growth factor receptor, as
well as putative biselective inhibitors of both receptor tyro-
sine kinases. However, this small-scale screen clearly high-
lights the complexity of identifying specific phenotypes for
screening.
Third, assays using 3D cell models are far less developed
with respect to imaging, analysis, quantification, and automa-
tion compared with established 2D methods. Confocal micros-
copy is the standard imaging tool for assessing cellular function
within 3D cell models; however, it is certainly limited in
throughput. Improvements in imaging modality, data acquisi-
tion throughput, and analysis tools are necessary for the wide
adoption of 3D cell cultures for screening.
Fourth, the predictive values of 3D cell cultures for drug
efficacy and toxicity need to be further determined and vali-
dated by using existing human data.145 Although data had
shown that the efficacy and toxicity of many drug mole-
cules obtained using 3D models are different from 2D cul-
tures, only a small set of these data confirmed that the
efficacy and toxicity of drugs in 3D models are close to the
clinical data.23,112,114,117
Fifth, regulatory authorities have yet to accept data
obtained from 3D cell models, such as organoids or organs-
on-chips, as a surrogate for preclinical animal testing. Partly
related to this is that historically, the assessment of new
technologies has been exceptionally slow (10 to 15 years)145
but more importantly is that these models often do not cap-
ture the full complexity of human organ function, such as
Fang and Eglen 13
lacking vascularization.146 In addition, organoid technolo-
gies face a common issue related to maturation.16
Nonetheless, 3D cell cultures have a bright future in drug
discovery and development. Three-dimensional cell cul-
tures would have enormous potential to model development
and disease, as advanced cell models under development
may fully capture the in vivo functions of organs and tis-
sues. Furthermore, the development of screening-compati-
ble 3D cell cultures would transform the drug discovery
process, as it becomes possible to obtain early the physio-
logically relevant efficacy and toxicity data. In addition, the
optimization of 3D cell cultures for scaling-up cell produc-
tion would improve quality, quantity, and efficacy, thus
making cells as therapeutics a reality.
Conclusion
A wide range of 3D cell culture technologies have been devel-
oped to address the need for continuously improving the pro-
ductivity of pharmaceutical R&D. Three-dimensional cell
cultures hold great potential as a tool for drug discovery—
ranging from disease modeling to target identification to
screening to lead identification—and as a new type of thera-
peutics/replacement therapy that may transform our lives.
Future developments in screening readily available 3D cell
models and assays, preclinically validated 3D cell models
for animal replacement, and functional, safe, and trans-
plantable 3D cell models will no doubt bring them closer to
reaching these potentials.
Acknowledgments
The authors appreciate valuable and constructive suggestions
from anonymous reviewers.
Declaration of Conflicting Interests
The authors declared no potential conflicts of interest with respect
to the research, authorship, and/or publication of this article.
Funding
The authors received no financial support for the research, author-
ship, and/or publication of this article.
References
1. Cukierman, E; Pankov, R; Stevens, D. R.; et al. Taking Cell-
Matrix Adhesions to the Third Dimension. Science 2001, 294,
1708–1712.
2. Bissell, M. J.; Rizki, A.; Mian, I, S. Tissue Architecture: The
Ultimate Regulator of Breast Epithelial Function. Curr. Opin.
Cell Biol. 2003, 15, 753–762.
3. Karlsson, H.; Fryknäs, M.; Larsson, R.; et al. Loss of Cancer
Drug Activity in Colon Cancer HCT-116 Cells during
Spheroid Formation in a New 3-D Spheroid Cell Culture
System. Exp. Cell Res. 2012, 318, 1577–1585.
4. Sodek, K. L.; Ringuette, M. J.; Brown, T. J. Compact
Spheroid Formation by Ovarian Cancer Cells Is Associated
with Contractile behavior and an Invasive Phenotype. Int. J.
Cancer 2009, 124, 2060–2070.
5. Kola, I.; Landis, J. Can the Pharmaceutical Industry Reduce
Attrition Rates? Nat. Rev. Drug Discov. 2004, 3, 711–715.
6. Paul, S. M.; Mytelka, D. S.; Dunwiddie, C. T.; et al. How to
Improve R&D Productivity: The Pharmaceutical Industry’s
Grand Challenge. Nat. Rev. Drug Discov. 2010, 9, 203–214.
7. Pammolli, F.; Magazzini, L.; Riccaboni, M. The Productivity
Crisis in Pharmaceutical R&D. Nat. Rev. Drug Discov. 2011,
10, 428–438.
8. Yamada, K. M.; Cukierman, E. Modeling Tissue
Morphogenesis and Cancer in 3D. Cell 2007, 130, 601–610.
9. Breslin, S.; O’Driscoll, L. Three-Dimensional Cell Culture:
The Missing Link in Drug Discovery. Drug Discov. Today
2013, 18, 240–248.
10. Sutherland, R. M.; Inch, W. R.; McCredie, J. A.; Kruuv, J.
A Multi-Component Radiation Survival Curve Using an In
Vitro Tumour Model. Int. J. Radiat. Biol. Relat. Stud. Phys.
Chem. Med. 1970, 18, 491–495.
11. Sutherland, R. M.; McCredie, J. A.; Inch, W. R. Growth of
Multicell Spheroids in Tissue Culture as a Model of Nodular
Carcinomas. J. Natl. Cancer Inst. 1971, 46, 113–120.
12. Vinci, M.; Gowan, S.; Boxall, F.; et al. Advances in
Establishment and Analysis of Three-Dimensional Tumor
Spheroid-Based Functional Assays for Target Validation and
Drug Evaluation. BMC Biol. 2012, 10, 29.
13. Tung, Y. C.; Hsiao, A. Y.; Allen, S. G.; et al. High-Throughput
3D Spheroid Culture and Drug Testing Using a 384 Hanging
Drop Array. Analyst 2011, 136, 473–478.
14. Youn, B. S.; Sen, A.; Behie, L. A.; et al. Scale-up of
Breast Cancer Stem Cell Aggregate Cultures to Suspension
Bioreactors. Biotechnol. Prog. 2006, 22, 801–810.
15. Yoshii, Y.; Waki, A.; Yoshida, K.; et al. The Use of
Nanoimprinted Scaffolds as 3D Culture Models to Facilitate
Spontaneous Tumor Cell Migration and Well-Regulated
Spheroid Formation. Biomaterials 2011, 32, 6052–6058.
16. Lancaster, M. A.; Knoblich, J. A. Organogenesis in a
Dish: Modeling Development and Disease Using Organoid
Technologies. Science 2014, 345, 124–125.
17. Willyard, C. Rise of the Organoids. Nature 2015, 523, 520–
522.
18. Shamir, E. R.; Ewald, A. J. Three-Dimensional Organotypic
Culture: Experimental Models of Mammalian Biology and
Disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 647–664.
19. Huch, M.; Koo, B. K. Modeling Mouse and Human
Development Using Organoid Cultures. Development 2015,
142, 3113–3125.
20. Antonica, F.; Kasprzyk, D. F.; Opitz, R.; et al. Generation
of Functional Thyroid from Embryonic Stem Cells. Nature
2012, 491, 66–71.
21. Greggio, C.; De Franceschi, F.; Figueiredo-Larsen, M.; et al.
Artificial Three-Dimensional Niches Deconstruct Pancreas
Development In Vitro. Development 2013, 140, 4452–4462.
22. Huch, M.; Dorrell, C.; Boj, S. F.; et al. In Vitro Expansion
of Single Lgr5+ Liver Stem Cells Induced by Wnt-Driven
Regeneration. Nature 2013, 494, 247–250.
14 SLAS Discovery
23. Takebe, T.; Sekine, K.; Enomura, M.; et al. Vascularized and
Functional Human Liver from an iPSC-Derived Organ Bud
Transplant. Nature 2013, 499, 481–484.
24. Barker, N.; Huch, M.; Kujala, P.; et al. Lgr5(+ve) Stem Cells
Drive Self-Renewal in the Stomach and Build Long-Lived
Gastric Units In Vitro. Cell Stem Cell 2010, 6, 25–36.
25. Stange, D. E.; Koo, B. K.; Huch, M.; et al. Differentiated
Troy+ Chief Cells Act as Reserve Stem Cells to Generate All
Lineages of the Stomach Epithelium. Cell 2013, 155, 357–
368.
26. Spence, J. R.; Mayhew, C. N.; Rankin, S. A.; et al. Directed
Differentiation of Human Pluripotent Stem Cells into
Intestinal Tissue In Vitro. Nature 2011, 470, 105–109.
27. Stevens, K. R.; Kreutziger, K. L.; Dupras, S. K.; et al.
Physiological Function and Transplantation of Scaffold-Free
and Vascularized Human Cardiac Muscle Tissue. Proc. Natl.
Acad. Sci. U.S.A. 2009, 106, 16568–16573.
28. Lancaster, M. A.; Renner, M.; Martin, C. A.; et al.
Cerebral Organoids Model Human Brain Development and
Microcephaly. Nature 2013, 501, 373–379.
29. Bredenkamp, N.; Ulyanchenko, S.; O’Neill, K. E.; et al. An
Organized and Functional Thymus Generated from FOXN1-
Reprogrammed Fibroblasts. Nat. Cell Biol. 2014, 16, 902–908.
30. Takasato, M.; Er, P. X.; Chiu, H. S.; et al. Kidney Organoids
from Human iPS Cells Contain Multiple Lineages and Model
Human Nephrogenesis. Nature 2015, 526, 564–568.
31. Freedman, B. S.; Brooks, C. R.; Lam, A. Q.; et al. Modelling
Kidney Disease with CRISPR-Mutant Kidney Organoids
Derived from Human Pluripotent Epiblast Spheroids. Nat.
Comm. 2015, 6, 8715.
32. Lee, J. H.; Bhang, D. H.; Beede, A.; et al. Lung Stem Cell
Differentiation in Mice Directed by Endothelial Cells via a
BMP4-NFATc1-Thrombospondin-1 Axis. Cell 2014, 156,
440–455.
33. Nakano, T.; Ando, S.; Takata, N.; et al. Self-Formation of
Optic Cups and Storable Stratified Neural Retina from Human
ESCs. Cell Stem Cell 2012, 10, 771–785.
34. Turner, D. A.; Baillie-Johnson, P.; Arias, A. Z. Organoids and
the Genetically Encoded Self-Assembly of Embryonic Stem
Cells. Bioessays 2015, 38, 181–191.
35. Kalabis, J.; Wong, G. S.; Vega, M. E.; et al. Isolation and
Characterization of Mouse and Human Esophageal Epithelial
Cells in 3D Organotypic Culture. Nat. Protoc. 2012, 7, 235–246.
36. Birgersdotter, A.; Sandberg, R.; Ernberg, I. Gene Expression
Perturbation In Vitro: A Growing Case for Three-Dimensional
(3D) Culture Systems. Semin. Cancer Biol. 2005, 15, 405–412.
37. Bryant, S. J.; Anseth, K. S. Hydrogel Properties Influence
ECM Production by Chondrocytes Photoencapsulated in
Poly(Ethylene Glycol) Hydrogels. J. Biomed. Mater. Res.
2002, 59, 63–72.
38. Tibbitt, M. W.; Anseth, K. S. Hydrogels as Extracellular
Matrix Mimics for 3D Cell Culture. Biotechnol. Bioeng.
2009, 103, 655–663.
39. Metters, A. T.; Anseth, K. S.; Bowman, C. N. Fundamental
Studies of a Novel, Biodegradable PEG-b-PLA Hydrogel.
Polymer 2000, 41, 3993–4004.
40. Lutolf, M. P.; Lauer-Fields, J. L.; Schmoekel, H. G.; et al.
Synthetic Matrix Metalloproteinase-Sensitive Hydrogels for
the Conduction of Tissue Regeneration: Engineering Cell-
Invasion Characteristics. Proc. Natl. Acad. Sci. U.S.A. 2003,
100, 5413–5418.
41. Nguyen, K. T.; West, J. L. Photopolymerizable Hydrogels
for Tissue Engineering Applications. Biomaterials 2002, 23,
4307–4314.
42. Benoit, D. S.; Schwartz, M. P.; Durney, A. R.; et al. Small
Functional Groups for Controlled Differentiation of Hydrogel-
Encapsulated Human Mesenchymal Stem Cells. Nat. Mater.
2008, 7, 816–823.
43. Murphy, W. L.; McDevitt, T. C.; Engler, A. J. Materials as
Stem Cell Regulators. Nat. Mater. 2014, 13, 547–557.
44. Engler, A. J.; Sen, S.; Sweeney, H. L.; et al. Matrix Elasticity
Directs Stem Cell Lineage Specification. Cell 2006, 126,
677–689.
45. Billiet, T.; Gevaert, E.; De Schryver, T.; et al. The 3D Printing
of Gelatin Methacrylamide Cell-Laden Tissue-Engineered
Constructs with High Cell Viability. Biomaterials 2014, 35,
49–62.
46. Ma, P. X.; Choi, J. W. Biodegradable Polymer Scaffolds with
Well-Defined Interconnected Spherical Pore Network. Tissue
Eng. 2001, 7, 23–33.
47. Sun, T.; Norton, D.; McKean, R. J.; et al. Development of a
3D Cell Culture System for Investigating Cell Interactions with
Electrospun Fibers. Biotechnol. Bioeng. 2007, 97, 1318–1328.
48. Loh, Q. L.; Choong, C. Three-Dimensional Scaffolds for
Tissue Engineering Applications: Role of Porosity and Pore
Size. Tissue Eng. Part B Rev. 2013, 19, 485–502.
49. Papadimitropoulos, A.; Piccinini, E.; Brachat, S.; et al.
Expansion of Human Mesenchymal Stromal Cells from Fresh
Bone Marrow in a 3D Scaffold-Based System under Direct
Perfusion. PLoS One 2014, 9, e102359.
50. Wise, J. K.; Yarin, A. L.; Megaridis, C. M.; et al. Chondrogenic
Differentiation of Human Mesenchymal Stem Cells on Oriented
Nanofibrous Scaffolds: Engineering the Superficial Zone of
Articular Cartilage. Tissue Eng. Part A 2009, 15, 913–921.
51. Khademhosseini, A.; Langer, R.; Borenstein, J.; et al.
Microscale Technologies for Tissue Engineering and Biology.
Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 2480–2487.
52. Whitesides, G. M. The Origins and the Future of Microfluidics.
Nature 2006, 442, 368–373.
53. Huh, D.; Hamilton, G. A.; Ingber, D. E. From 3D Cell Culture
to Organs-on-Chips. Trends Cell Biol. 2011, 21, 745–754.
54. Bhatia, S. N.; Ingber, D. E. Microfluidic Organs-on-Chip.
Nat. Biotechnol. 2014, 32, 760–772.
55. Li, L.; Fukunaga-Kalabis, M.; Herlyn, M. The Three-
Dimensional Human Skin Reconstruct Model: A Tool to
Study Normal Skin and Melanoma Progression. J. Vis. Exp.
2011, 54, 2937.
56. Huh, D.; Matthews, B. D.; Mammoto, A.; et al. Reconstituting
Organ-Level Lung Functions on a Chip. Science 2010, 328,
1662–1668.
57. Huh, D.; Leslie, D. C.; Matthews, B. D.; et al. A Human
Disease Model of Drug Toxicity-Induced Pulmonary Edema
in a Lung-on-a-Chip Microdevice. Sci. Transl. Med. 2012, 4,
159ra147.
58. Benam, K. H.; Novak, R.; Nawroth, J.; et al. Matched-
Comparative Modeling of Normal and Diseased Human
Fang and Eglen 15
Airway Responses Using a Microengineered Breathing Lung
Chip. Cell Syst. 2016, 3, 456–466.
59. Song, J. W.; Cavnar, S. P.; Walker, A. C.; et al. Microfluidic
Endothelium for Studying the Intravascular Adhesion of
Metastatic Breast Cancer Cells. PLoS One 2009, 4, e5756.
60. Zhang, Y. S.; Aleman, J.; Arneri, A.; et al. From Cardiac
Tissue Engineering to Heart-on-a-Chip: Beating Challenges.
Biomed. Mater. 2015, 10, 034006.
61. Kim, S. B.; Bae, H.; Cha, J. M.; et al. A Cell-Based Biosensor
for Real-Time Detection of Cardiotoxicity Using Lensfree
Imaging. Lab Chip 2011, 11, 1801–1807.
62. Cvetkovic, C.; Raman, R.; Chan, V.; et al. Three-
Dimensionally Printed Biological Machines Powered by
Skeletal Muscle. Proc. Natl. Acad. Sci. U.S.A. 2014, 111,
10125–10130.
63. LeCluyse, E. L.; Witek, R. P.; Andersen, M. E.; et al.
Organotypic Liver Culture Models: Meeting Current
Challenges in Toxicity Testing. Crit. Rev. Toxicol. 2012, 42,
501–548.
64. Baudoin, R.; Prot, J. M.; Nicolas, G.; et al. Evaluation of
Seven Drug Metabolisms and Clearances by Cryopreserved
Human Primary Hepatocytes Cultivated in Microfluidic
Biochips. Xenobiotica 2013, 43, 140–152.
65. Chao, P.; Maguire, T.; Novik, E.; et al. Evaluation of a
Microfluidic Based Cell Culture Platform with Primary
Human Hepatocytes for the Prediction of Hepatic Clearance
in Human. Biochem. Pharmacol. 2009, 78, 625–632.
66. Gao, D.; Liu, H.; Lin, J. M.; et al. Characterization of Drug
Permeability in Caco-2 Monolayers by Mass Spectrometry on
a Membrane-Based Microfluidic Device. Lab Chip 2013, 13,
978–985.
67. Domansky, K.; Inman, W.; Serdy, J.; et al. Perfused Multiwell
Plate for 3D Liver Tissue Engineering. Lab Chip 2010, 10,
51–58.
68. Sarkar, U.; Rivera-Burgos, D.; Large, E. M.; et al. Metabolite
Profiling and Pharmacokinetic Evaluation of Hydrocortisone
in a Perfused Three-Dimensional Human Liver Bioreactor.
Drug Metab. Dispos. 2015, 43, 1091–1099.
69. Murphy, S. V.; Atala, A 3D Bioprinting of Tissues and
Organs. Nat. Biotechnol. 2014, 32, 773–785.
70. Ingber, D. E.; Mow, V. C.; Butler, D.; et al. Tissue Engineering
and Developmental Biology: Going Biomimetic. Tissue Eng.
2006, 12, 3265–3283.
71. Derby, B. Printing and Prototyping of Tissues and Scaffolds.
Science 2012, 338, 921–926.
72. Mironov, V.; Visconti, R. P.; Kasyanov, V.; et al. Organ
Printing: Tissue Spheroids as Building Blocks. Biomaterials
2009, 30, 2164–2174.
73. Kelm, J. M.; Lorber, V.; Snedeker, J. G.; et al. A Novel
Concept for Scaffold-Free Vessel Tissue Engineering: Self-
assembly of Microtissue Building Blocks. J. Biotechnol.
2010, 148, 46–55.
74. Zhang, Y. S.; Yue, K.; Aleman, J.; et al. 3D Bioprinting for
Tissue and Organ Fabrication. Ann. Biomed. Eng. 2017, 45,
148–163.
75. Peng, W.; Unutmaz, D.; Ozbolat, I. T. Bioprinting towards
Physiologically Relevant Tissue Models for Pharmaceutics.
Trends Biotechnol. 2016, 34, 722–732.
76. Hughes, J. P.; Rees, S.; Kalindjian, S. B.; et al. Principles of
Early Drug Discovery. Br. J. Pharmacol. 2011, 162, 1239–
1249.
77. Rask-Andersen, M.; Almén, M. S.; Schiöth, H. B. Trends
in the Exploitation of Novel Drug Targets. Nat. Rev. Drug
Discov. 2011, 10, 579–590.
78. Tanner, K.; Gottesman, M. M. Beyond 3D Culture Models of
Cancer. Sci. Transl. Med. 2015, 7, 283ps9 .
79. Rizki, A.; Weaver, V. M.; Lee, S. Y.; et al. A Human Breast
Cell Model of Preinvasive to Invasive Transition. Cancer Res.
2008, 68, 1378–1387.
80. Paszek, M. J.; Zahir, N.; Johnson, K. R.; et al. Tensional
Homeostasis and the Malignant Phenotype. Cancer Cell
2005, 8, 241–254.
81. Drost, J.; van Jaarsveld, R. H.; Ponsioen, B.; et al. Sequential
Cancer Mutations in Cultured Human Intestinal Stem Cells.
Nature 2015, 521, 43–47.
82. Weiswald, L. B.; Bellet, D.; Dangles-Marie, V. Spherical
Cancer Models in Tumor Biology. Neoplasia 2015, 17, 1–15.
83. Dieter, S. M.; Ball, C. R.; Hoffmann, C. M.; et al. Distinct
Types of Tumor-Initiating Cells form Human Colon Cancer
Tumors and Metastases. Cell Stem Cell 2011, 9, 357–365.
84. Hanahan, D.; Weinberg, R. A. Hallmarks of Cancer: The Next
Generation. Cell 2011, 144, 646–674.
85. Kniazeva, E.; Putnam, A. J. Endothelial Cell Traction and
ECM Density Influence Both Capillary Morphogenesis and
Maintenance in 3-D. Am. J. Physiol. Cell Physiol. 2009, 297,
C179–C187.
86. Blacher, S.; Erpicum, C.; Lenoir, B.; et al. Cell Invasion in the
Spheroid Sprouting Assay: A Spatial Organization Analysis
Adaptable to Cell Behaviour. PLoS One 2014, 9, e97019.
87. Ghosh, S.; Joshi, M. B.; Ivanov, D.; et al. Use of Multicellular
Tumor Spheroids to Dissect Endothelial Cell–Tumor Cell
Interactions: A Role for T-Cadherin in Tumor Angiogenesis.
FEBS Lett. 2007, 581, 4523–4528.
88. Febles, N. K.; Ferrie, A. M.; Fang, Y. Label-Free Single Cell
Quantification of the Invasion of Spheroidal Colon Cancer
Cells through 3D Matrigel. Anal. Chem. 2014, 86, 8842–
8849.
89. Chandrasekaran, S.; Deng, H.; Fang, Y. PTEN Deletion
Potentiates Invasion of Colorectal Cancer Spheroidal Cells
through 3D Matrigel. Integr. Biol. (Camb.) 2015, 7, 324–334.
90. Finkbeiner, S. R.; Zeng, X. L.; Utama, B.; et al. Stem Cell-
Derived Human Intestinal Organoids as an Infection Model
for Rotaviruses. Mbio. 2012, 3, e00159-12.
91. Castellanos-Gonzalez, A.; Cabada, M. M.; Nichols, J.; et al.
Human Primary Intestinal Epithelial Cells as an Improved In
Vitro Model for Cryptosporidium parvum Infection. Infect.
Immun. 2013, 81, 1996–2001.
92. Yeung, T. M.; Gandhi, S. C.; Wilding, J. L.; et al. Cancer
Stem Cells from Colorectal Cancer Derived Cell Lines. Proc.
Natl. Acad. Sci. U.S.A. 2010, 107, 3722–3727.
93. Onuma, K.; Ochiai, M.; Orihashi, K.; et al. Genetic
Reconstitution of Tumorigenesis in Primary Intestinal Cells.
Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 11127–11132.
94. Bigorgne, A. E.; Farin, H. F.; Lemoine, R.; et al. TTC7A
Mutations Disrupt Intestinal Epithelial Apicobasal Polarity. J.
Clin. Invest. 2013, 124, 328–337.
16 SLAS Discovery
95. Dekkers, J. F.; Wiegerinck, C. L.; de Jonge, H. R.; et al.
A Functional CFTR Assay Using Primary Cystic Fibrosis
Intestinal Organoids. Nat. Med. 2013, 19, 939–945.
96. Matano, M.; Date, S.; Shimokawa, M.; et al. Modeling
Colorectal Cancer Using CRISPR-Cas9-Mediated Engineering
of Human Intestinal Organoids. Nat. Med. 2015, 21, 256–262.
97. Simon, G. M.; Niphakis, M. J.; Cravatt, B. F. Determining
Target Engagement in Living Systems. Nat. Chem. Biol.
2013, 9, 200–205.
98. Ghosh, S.; Spagnoli, G. C.; Martin, I.; et al. Three-
Dimensional Culture of Melanoma Cells Profoundly Affects
Gene Expression Profile: A High Density Oligonucleotide
Array Study. J. Cell Physiol. 2005, 204, 522–531.
99. Barbone, D.; Van Dam, L.; Follo, C.; et al. Analysis of Gene
Expression in 3D Spheroids Highlights a Survival Role for
ASS1 in Mesothelioma. PLoS One 2016, 11, e0150044.
100. Swinney, D. C.; Anthony, J. How Were New Medicines
Discovered? Nat. Rev. Drug Discov. 2011, 10, 507–519.
101. Feng, Y.; Mitchison, T. J.; Bender, A.; et al. Multi-Parameter
Phenotypic Profiling: Using Cellular Effects to Characterize
Small-Molecule Compounds. Nat. Rev. Drug Discov. 2009,
8, 567–578.
102. Fang, Y. Label-Free Cell Phenotypic Drug Discovery.
Comb. Chem. High Throughput Screen. 2014, 17, 566–578.
103. Fang, Y. Label-Free Chemical and Phenotypic Profiling of
Living Cells. Sci. Lett. 2015, 4, 156.
104. Esch, E. W.; Bahinski, A.; Huh, D. Organs-on-Chips at the
Frontiers of Drug Discovery. Nat. Rev. Drug Discov. 2015,
14, 248–260.
105. Senkowski, W.; Zhang, X.; Olofsson, M. H.; et al. Three-
Dimensional Cell Culture-Based Screening Identifies the
Anthelmintic Drug Nitazoxanide as a Candidate for Treatment
of Colorectal Cancer. Mol. Cancer Ther. 2015, 14, 1504–1516.
106. Wenzel, C.; Riefke, B.; Gründemann, S.; et al. 3D High-
Content Screening for the Identification of Compounds That
Target Cells in Dormant Tumor Spheroid Regions. Exp. Cell
Res. 2014, 323, 131–143.
107. Kenny, H. A.; Lal-Nag, M.; White, E. A.; et al. Quantitative
High Throughput Screening Using a Primary Human Three-
Dimensional Organotypic Culture Predicts In Vivo Efficacy.
Nat. Commun. 2015, 6, 6220.
108. Hay, M.; Thomas, D. W.; Craighead, J. L.; et al. Clinical
Development Success Rates for Investigational Drugs. Nat.
Biotechnol. 2014, 32, 40–51.
109. Mehta, G.; Hsiao, A. Y.; Ingram, M.; et al. Opportunities
and Challenges for Use of Tumor Spheroids as Models to
Test Drug Delivery and Efficacy. J. Control Release 2012,
164, 192–204.
110. Hickman, J. A.; Graeser, R.; de Hoogt, R.; et al. IMI
PREDECT Consortium. Three-Dimensional Models
of Cancer for Pharmacology and Cancer Cell Biology:
Capturing Tumor Complexity In Vitro/Ex Vivo. Biotechnol.
J. 2014, 9, 1115–1128.
111. Fatehullah, A.; Tan, S. H.; Barker, N. Organoids as an In
Vitro Model of Human Development and Disease. Nat. Cell
Biol. 2015, 8, 246–254.
112. Halfter, K.; Hoffmann, O.; Ditsch, N.; et al. Testing
Chemotherapy Efficacy in HER2 Negative Breast Cancer Using
Patient-Derived Spheroids. J. Transl. Med. 2016, 14, 112.
113. Tong, J. G.; Valdes, Y. R.; Barrett, J. W.; et al. Evidence for
Differential Viral Oncolytic Efficacy in an In Vitro Model
of Epithelial Ovarian Cancer Metastasis. Mol. Ther. Oncol.
2015, 2, 15013.
114. Aref, A. R.; Huang, R. Y.; Yu, W.; et al. Screening
Therapeutic EMT Blocking Agents in a Three-Dimensional
Microenvironment. Integr. Biol. (Camb.). 2013, 5, 381–389.
115. Wilke, R. A.; Lin, D. W.; Roden, D. M.; et al. Identifying
Genetic Risk Factors for Serious Adverse Drug Reactions:
Current Progress and Challenges. Nat. Rev. Drug Discov.
2007, 6, 904–916.
116. Schwartz, M. P.; Hou, Z.; Propson, N. E.; et al. Human
Pluripotent Stem Cell-Derived Neural Constructs for
Predicting Neural Toxicity. Proc. Natl. Acad. Sci. U.S.A.
2015, 112, 12516–12521.
117. Gunness, P.; Mueller, D.; Shevchenko, V.; et al. 3D
Organotypic Cultures of Human HepaRG Cells: A Tool for
In Vitro Toxicity Studies. Toxicol. Sci. 2013, 133, 67–78.
118. Bell, C. C.; Hendriks, D. F.; Moro, S. M.; et al.
Characterization of Primary Human Hepatocyte Spheroids
as a Model System for Drug-Induced Liver Injury, Liver
Function and Disease. Sci. Rep. 2016, 6, 25187.
119. Lee, J. B.; Sung, J. H. Organ-on-a-Chip Technology and
Microfluidic Whole-Body Models for Pharmacokinetic Drug
Toxicity Screening. Biotechnol. J. 2013, 8, 1258–1266.
120. Chan, C. Y.; Huang, P. H.; Guo, F.; et al. Accelerating Drug
Discovery via Organs-on-Chips. Lab Chip 2013, 13, 4697–
4710.
121. Wikswo, J. P.; Curtis, E. L.; Eagleton, Z. E.; et al. Scaling
and Systems Biology for Integrating Multiple Organs-on-a-
Chip. Lab Chip 2013, 13, 3496–3511.
122. Baraniak, P. R.; McDevitt, T. C. Scaffold-Free Culture of
Mesenchymal Stem Cell Spheroids in Suspension Preserves
Multilineage Potential. Cell Tissue Res. 2012, 347, 701–711.
123. Galipeau, J. The Mesenchymal Stromal Cells Dilemma—
Does a Negative Phase III Trial of Random Donor
Mesenchymal Stromal Cells in Steroid-Resistant Graft-
Versus-Host Disease Represent a Death Knell or a Bump in
the Road? Cytotherapy 2013, 15, 2–8.
124. Tongers, J.; Losordo, D. W.; Landmesser, U. Stem and
Progenitor Cell-Based Therapy in Ischaemic Heart Disease:
Promise, Uncertainties, and Challenges. Eur. Heart J. 2011,
32, 1197–1206.
125. Park, E.; Patel, A. N. Changes in the Expression Pattern of
Mesenchymal and Pluripotent Markers in Human Adipose-
Derived Stem Cells. Cell Biol. Int. 2010, 34, 979–984.
126. Baer, P. C.; Griesche, N.; Luttmann, W.; et al. Human
Adipose-Derived Mesenchymal Stem Cells In Vitro:
Evaluation of an Optimal Expansion Medium Preserving
Stemness. Cytotherapy 2010, 12, 96–106.
127. Cesarz, Z.; Tamama, K. Spheroid Culture of Mesenchymal
Stem Cells. Stem Cells Int. 2016, 2016, 9176357.
128. Tsai, A. C.; Liu, Y.; Yuan, X.; et al. Compaction, Fusion,
and Functional Activation of Three-Dimensional Human
Mesenchymal Stem Cell Aggregate. Tissue Eng. A 2015, 21,
1705–1719.
129. Ruiz, S. A.; Chen, C. S. Emergence of Patterned Stem Cell
Differentiation within Multicellular Structures. Stem Cells
2008, 26, 2921–2927.
Fang and Eglen 17
130. Potapova, I. A.; Gaudette, G. R.; Brink, P. R.; et al.
Mesenchymal Stem Cells Support Migration, Extracellular
Matrix Invasion, Proliferation, and Survival of Endothelial
Cells In Vitro. Stem Cells 2007, 25, 1761–1768.
131. Xu, Y.; Shi, T.; Xu, A.; et al. 3D Spheroid Culture
Enhances Survival and Therapeutic Capacities of MSCs
Injected into Ischemic Kidney. J. Cell Mol. Med. 2016,
20, 1203–1213.
132. Henry, E.; Cores, J.; Hensley, M. T.; et al. Adult Lung Spheroid
Cells Contain Progenitor Cells and Mediate Regeneration in
Rodents with Bleomycin-Induced Pulmonary Fibrosis. Stem
Cells Transl. Med. 2015, 4, 1265–1274.
133. Schulz, T. C. Concise Review: Manufacturing of Pancreatic
Endoderm Cells for Clinical Trials in Type 1 Diabetes. Stem
Cells Transl. Med. 2015, 4, 927–931.
134. Taguchi, A.; Kaku, Y.; Ohmori, T.; et al. Redefining the In
Vivo Origin of Metanephric Nephron Progenitors Enables
Generation of Complex Kidney Structures from Pluripotent
Stem Cells. Cell Stem Cell 2014, 14, 53–67.
135. Fordham, R. P.; Yui, S.; Hannan, N. R.; et al. Transplantation
of Expanded Fetal Intestinal Progenitors Contributes to
Colon Regeneration after Injury. Cell Stem Cell 2013, 13,
734–744.
136. Yui, S.; Nakamura, T.; Sato, T.; et al. Functional Engraftment
of Colon Epithelium Expanded In Vitro from a Single Adult
Lgr5+ Stem Cell. Nat. Med. 2012, 18, 618–623.
137. Atala, A.; Bauer, S. B.; Soker, S.; et al. Tissue-Engineered
Autologous Bladders for Patients Needing Cystoplasty.
Lancet 2006, 367, 1241–1246.
138. Onoe, H.; Okitsu, T.; Itou, A.; et al. Metre-Long Cell-Laden
Microfibres Exhibit Tissue Morphologies and Functions.
Nat. Mater. 2013, 12, 584–590.
139. Munoz-Abraham, A. S.; Rodriguez-Davalos, M. I.; Bertacco,
A.; et al. 3D Printing of Organs for Transplantation: Where
Are We and Where Are We Heading? Curr. Transplant Rep.
2016, 3, 93–99.
140. Norotte, C.; Marga, F. S.; Niklason, L. E.; et al. Scaffold-
Free Vascular Tissue Engineering Using Bioprinting.
Biomaterials 2009, 30, 5910–5917.
141. Visconti, R. P.; Kasyanov, V.; Gentile, C.; et al. Towards
Organ Printing: Engineering an Intra-Organ Branched
Vascular Tree. Expert Opin. Biol. Ther. 2010, 10, 409–420.
142. Miller, J. S.; Stevens, K. R.; Yang, M. T.; et al. Rapid Casting
of Patterned Vascular Networks for Perfusable Engineered
Three-Dimensional Tissues. Nat. Mater. 2012, 11, 768–774.
143. Li, L.; Zhou, Q.; Voss, T. C.; et al. High-Throughput
Imaging: Focusing in on Drug Discovery in 3D. Methods
2016, 96, 97–102.
144. Booij, T. H.; Klop, M. J.; Yan, K.; et al. Development of a
3D Tissue Culture-Based High-Content Screening Platform
That Uses Phenotypic Profiling to Discriminate Selective
Inhibitors of Receptor Tyrosine Kinases. J. Biomol. Screen.
2016, 21, 912–922.
145. Dambach, D. M.; Uppal, H. Improving Risk Assessment.
Sci. Transl. Med. 2012, 4, 159ps22.
146. Bahinski, A.; Horland, R.; Huh, D.; et al. The promise and
potential of “organs-on-chips” as preclinical models. Appl.
In Vitro Toxicol. 2015, 1, 235–242.
... Due to the physiological similarity to in vivo, three-dimensional (3D) cell culture technologies, including cell spheroids, organoids, 3D bioprinted tissue constructs, and microphysiological systems, have been rapidly evolving (Fang and Eglen 2017;Larson 2015). There is growing evidence that 3D in vitro tumor models enable unprecedented phenotypic analysis by mimicking tissue structure and function and emulating in vivo characteristics compared two-dimensional (2D) culture of cell monolayers (Jung et al. 2017;Lee et al. 2021;Song et al. 2018). ...
Article
Full-text available
Recent advancements in three‐dimensional (3D) cell culture technologies, such as cell spheroids, organoids, and 3D bioprinted tissue constructs, have significantly improved the physiological relevance of in vitro models. These models better mimic tissue structure and function, closely emulating in vivo characteristics and enhancing phenotypic analysis, critical for basic research and drug screening in personalized cancer therapy. Despite their potential, current 3D cell culture platforms face technical challenges, which include user‐unfriendliness in long‐term dynamic cell culture, incompatibility with rapid cell encapsulation in biomimetic hydrogels, and low throughput for compound screening. To address these issues, we developed a 144‐pillar plate with sidewalls and slits (144PillarPlate) and a complementary 144‐perfusion plate with perfusion wells and reservoirs (144PerfusionPlate) for dynamic 3D cell culture and predictive compound screening. To accelerate biomimetic tissue formation, small Hep3B liver tumor spheroids suspended in alginate were printed and encapsulated on the 144PillarPlate rapidly by using microsolenoid valve‐driven 3D bioprinting technology. The microarray bioprinting technology enabled precise and rapid loading of small spheroids in alginate on the pillar plate, facilitating reproducible and scalable formation of large tumor spheroids with minimal manual intervention. The bioprinted Hep3B spheroids on the 144PillarPlate were dynamically cultured in the 144PerfusionPlate and tested with anticancer drugs to measure drug effectiveness and determine the concentration required to inhibit 50% of the cell viability (IC50 value). The perfusion plate enabled the convenient dynamic culture of tumor spheroids and facilitated the dynamic testing of anticancer drugs with increased sensitivity. It is envisioned that the integration of microarray bioprinting of tumor spheroids onto the pillar plate, along with dynamic 3D cell culture in the perfusion plate, could more accurately replicate tumor microenvironments. This advancement has the potential to enhance the predictive drug screening process in personalized cancer therapy significantly.
... Three-dimensional (3D) tissue culture has become an important bridge between in vitro and in vivo models in various biological research fields [1,2]. Three-dimensional structures provide in vivo-like conditions that are beneficial for studying cell physiology and conducting high-throughput drug discovery and screening [3][4][5]. In 3D clusters like ...
Article
Full-text available
Three-dimensional (3D) tissue culture models provide in vivo-like conditions for studying cell physiology. This study aimed to examine the efficiency of pyramidal microwell geometries in microfluidic devices on spheroid formation, cell growth, viability, and differentiation in mouse embryonic stem cells (mESCs). The static culture using the hanging drop (HD) method served as a control. The microfluidic chips were fabricated to have varying pyramidal tip angles, including 66°, 90°, and 106°. From flow simulations, when the tip angle increased, streamline distortion decreased, resulting in more uniform flow and a lower velocity gradient near the spheroids. These findings demonstrate the significant influence of microwell geometry on fluid dynamics. The 90° microwells provide optimal conditions, including uniform flow and reduced shear stress, while maintaining the ability for waste removal, resulting in superior spheroid growth compared to the HD method and other microwell designs. From the experiments, by Day 3, spheroids in the 90° microwells reached approximately 400 µm in diameter which was significantly larger than those in the 66° microwells, 106° microwells, and HD cultures. Brachyury gene expression in the 90° microwells was four times higher than the HD method, indicating enhanced mesodermal differentiation essential for cardiac differentiation. Immunofluorescence staining confirmed cardiomyocyte differentiation. In conclusion, microwell geometry significantly influences 3D cell culture outcomes. The pyramidal microwells with a 90° tip angle proved most effective in promoting spheroid growth and cardiac differentiation of mESC differentiation, providing insights for optimizing microfluidic systems in tissue engineering and regenerative medicine.
... The main advantage of developing research using cell culture assays is based on the possibility of investigating the specific role of receptors, cell signaling molecules, enzymes, and mediators by pharmacological blockade (4). Such experimental designs are useful for understanding disease mechanisms as well as biomaterials' effects on living cells (5)(6)(7). ...
Article
Full-text available
Dimethyl sulfoxide (DMSO) is widely used as an adjuvant in dissolving insoluble compounds in an aqueous medium; however, it can induce significant molecular changes in cells. The possible damages may occur obeying a tissue-specific profile, and the effect on human apical papilla cells (hAPC) remains unknown. Therefore, this study aimed to evaluate DMSO effects on the viability and mineralization activity in hAPC cultures in vitro and to establish standards of maximum concentrations for its use in laboratory routines. hAPCs were cultured, plated, and maintained in media containing increasing concentrations of Dimethyl sulfoxide (0.1%, 0.5%, 1%, 5%, and 10%) for 24 h, 48 h, 72 h, and 7 days. At each time point, the cells were subjected to the MTT assay. The Alizarin red S staining assay was performed to evaluate the osteo/odontogenic mineralization potential of hAPC DMSO-exposed (0.1%, 0.5%, and 1%) in the 21-day time-point. Statistical analysis was performed using one-way analysis of variance followed by Tukey's post hoc test (p<0.05). In general, the 5% and 10% DMSO concentrations were shown to be cytotoxic for hAPC at all analyzed time points, and the hAPC DMSO-stimulated presented higher osteo/odontogenic mineralization potential. Therefore, the 5% and 10% DMSO concentrations should be avoided, and the mineralization activity assay should be carefully designed in order to avoid biases at in vitro assays using hAPC cultures.
... The morphology of cells growth becomes more diverse in 3D model culture and resembles the diversity in vivo tissues [39,43,44]. Additionally, physiological interactions between cells and the ECM are replicated in a 3D model culture, which more accurately mimics the growth environment within the body [45,46]. Compared to 2D cell culture, 3D model culture better simulate signaling between cells. ...
Article
Full-text available
The aim of this study was to develop a three-dimensional (3D) cell model in order to evaluate the effectiveness of a traditional Chinese medicine decoction in the treatment of arthritis. Chondrocytes (ATDC5) and osteoblasts (MC3T3-E1) were 3D printed separately using methacryloyl gelatin (GelMA) hydrogel bioinks to mimic the natural 3D cell environment. Both cell types showed good biocompatibility in GelMA. Lipopolysaccharide (LPS) was added to the cell models to create inflammation models, which resulted in increased expression of inflammatory factors IL-1β, TNF-α, iNOS, and IL-6, and decreased expression of cell functional genes such as Collagen II (COLII), transcription factor SOX-9 (Sox9), Aggrecan, alkaline phosphatase (ALP), RUNX family transcription factor 2 (Runx2), Collagen I (COLI), Osteopontin (OPN), and bone morphogenetic protein-2 (BMP-2). The created inflammation model was then used to evaluate the effectiveness of Dangguiniantongtang (DGNT) decoctions. The results showed that DGNT reduced the expression of inflammatory factors and increased the expression of functional genes in the cell model. In summary, this study established a 3D cell model to assess the effectiveness of traditional Chinese medicine (TCM) decoctions, characterized the gene expression profile of the inflammatory state model, and provided a practical reference for future research on TCM efficacy evaluation for arthritis treatment.
Article
Pancreatic islet transplantation holds great potential as a therapeutic approach for treating type 1 diabetes mellitus (T1D). However, large islets suffer from hypoxia due to the limited diffusion distance of oxygen, leading to cell loss. Therefore, smaller spheroids are needed for better transplantation outcomes. This study aims to develop a method for forming highly functional islet spheroids using glycol chitosan (GC) derivatives, such as N-acetylated glycol chitosan (AGC) and N-hexanoyl glycol chitosan (HGC). Thermogelling polymers were produced by performing N-acylation of GC using the correspondingly carboxylic anhydrides. Islet spheroids were formed using a dual application with AGC-coated plates and HGC gelation. The AGC solution was applied to the plate for coating and evenly distributed using a 1 mL syringe. Then, the HGC encapsulated with islet single cells was cultured on top of it. Spheroid viability and functionality were evaluated using CCK-8 assay and glucose-stimulated insulin secretion assay. The aqueous solutions of AGC (4%, w/v) and HGC (36% hexanoylation) (2%, w/v) demonstrated a sol–gel transition temperature around 37 °C, suitable for the physiological environment. These polymers also showed no cytotoxicity to intact islets. Islet single cells were cultured on HGC gels with varying degrees of hexanoylation (DH) values, where higher DH values led to smaller and more uniform spheroids. The resulting spheroids formed on AGC-coated plates and HGC36 gelation were smaller and more uniform than those formed on untreated plates. These spheroids exhibited significantly improved glucose responsiveness, with superior insulin secretion. The optimized method using AGC and HGC offers a more efficient way to produce smaller, uniform, and functional spheroids.
Article
The increasing prevalence of neurological, neurodegenerative and psychiatric diseases makes it necessary to create appropriate and informative models to study the molecular mechanisms of their pathogenesis and search for promising approaches to treat them. 2D neuron cultures do not display the full complexity of intercellular interactions in the nervous system, and the brains of even primitive mammals are too complex and not always suitable for modeling human brain pathologies. The missing link between 2D-cultures and in vivo models are organotypic 3D-cultures. Such models include surviving brain slices, self-organizing 3D-cultures (neurospheres, brain organoids, assembloids) and brain-on-a-chip cultures. These 3D-models make it possible to reproduce not only the cellular composition of the brain, but to a large extent its architectonics or even to simulate its interactions with other organs and tissues within the body. Their application has already made it possible to obtain data on some molecular mechanisms involved in the development of hereditary, inflammatory and neurodegenerative diseases of the central nervous system, as well as to propose and test some approaches to their treatment. This literature review discusses the possibilities, limitations and the most striking examples of the use of these organotypic brain models. The use of 3D human and animal brain cultures opens unprecedented perspectives for the development of neurobiology, molecular psychiatry and neurology. The high potential and flexibility of organotypic models, the increasing need for modeling various brain pathologies and the striving to establish causal mechanisms of disease allow us to assert that in the very near future 3D-models will experience rapid growth, development and widespread use.
Article
Full-text available
In recent years, three-dimensional (3D) spheroid technology has emerged as a crucial instrument in the realm of cancer research, offering a near-physiological model for probing human cancers. By faithfully replicating organ architecture and functionality, spheroids furnish a versatile platform for addressing a spectrum of clinical and biomedical inquiries, encompassing pharmacology and disease pathophysiology. Their distinct advantage over conventional two-dimensional (2D) cell cultures lies in their capacity to emulate the 3D extracellular microenvironment and attributes characteristic of solid tumors, including architectural intricacies, gene expression profiles, and secretion of soluble mediators. Derived effectively from both normal and malignant patient tissues, spheroids facilitate the modeling of cancer progression, mutation dynamics, and carcinogenesis pathways. Moreover, spheroid technology expedites drug screening processes and personalized therapeutic interventions. Although challenges persist in accurately recapitulating the immune system within spheroid models, the co-culturing of spheroids with lymphocytes holds significant promise for immunotherapy applications. This comprehensive examination outlines diverse methodologies for establishing and characterizing spheroids, highlighting their extensive utilization in oncology. The manuscript underscores the immense promise of 3D spheroids in cancer research, paving the way for an uprising in the understanding and management of cancer, with abundant opportunities for further investigation and progress in treatment approaches.
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas’ functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevant in vitro human models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally, α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.
Article
Glioblastoma is a highly malignant brain cancer with a poor survival rate. Increasing evidence demonstrates the anticancer activity, including anti-glioma activity, of indomethacin (IND), a non-steroidal anti-inflammatory drug. However, due to the IND's poor aqueous solubility, nano-based drug delivery systems, especially gold nanoparticles (AuNPs), are great tools for increasing solubility and therapeutic efficacy. Herein, glutathione (GSH)-coated folic acid (FA)-modified AuNPs were used for the first time to generate IND-loaded AuNPs (55 nm), which were successfully synthesized according to DLS, TEM, FTIR, NMR, and TGA results. IND/AuNPs were found to have spherical morphology, nanoscale particle size, narrow size distribution, and good stability. Fluorescence and confocal imaging demonstrated that the nanoparticles penetrated folate receptor (FR)+ U-87MG human glioblastoma monolayer and sphere-forming cells. Remarkably, short-term exposure (4h) to IND/AuNPs significantly increased IND cytotoxicity in U-87MG cells after post-44h and -68h (>35- and >120-fold, respectively). Even against prolonged exposure of cells to IND for 24h, 48h, and 72h, IND/AuNP treatment revealed a marked result: glioma proliferation slowed by 7.38-fold, 6.8-fold, and 17-fold, respectively. No significant effect was observed on the FR− cell lines. The increased antitumoral activity was accompanied by efficient increased apoptosis in glioblastoma cells due to the IND/AuNP treatments. Moreover, compared to free-drug and control groups, IND/AuNP treatments markedly reduced glioblastoma growth in 3D spheroids (in vitro system that mimics in vivo tumors). Therefore, these findings suggest that the new spherical IND/Au-GSH-FA NP conjugate has the potential to be a beneficial therapeutic agent in glioblastoma therapy by targeting FRs.
Article
Full-text available
Cancer is a collection of illnesses characterized by aberrant cellular proliferation that can infiltrate or metastasize to distant anatomical sites, posing a notable threat to human well‐being due to its substantial morbidity and death rates worldwide. The potential of plant‐derived natural compounds as anticancer medicines has been assessed owing to their favorable attributes of few side effects and significant antitumor activity. Mangrove plants and their derived compounds have been scientifically shown to exhibit many significant beneficial biological activities, such as anti‐inflammatory, immunomodulatory, antioxidant, neuroprotective, cardioprotective, and hepatoprotective properties. This study summarized mangrove plants and their derived compounds as potential anticancer agents, with an emphasis on the underlying molecular mechanisms. To explore this, we gathered data on the preclinical (in vivo and in vitro) anticancer effects of mangrove plants and their derived compounds from reputable literature spanning 2000 to 2023. We conducted thorough searches in various academic databases, including PubMed, ScienceDirect, Wiley Online, SpringerLink, Google Scholar, Scopus, and the Web of Science. The results demonstrated that mangrove plants and their derived compounds have promising anticancer properties in preclinical pharmacological test systems through various molecular mechanisms, including induction of oxidative stress and mitochondrial dysfunction, cytotoxicity, genotoxicity, cell cycle arrest, apoptosis, autophagy, antiproliferative, antimetastatic, and other miscellaneous actions. Upon thorough observation of the pertinent information, it is suggested that mangrove plants and their derived chemicals may serve as a potential lead in the development of novel drugs for cancer therapy.
Article
Full-text available
3D tissue cultures provide a more physiologically relevant context for the screening of compounds, compared with 2D cell cultures. Cells cultured in 3D hydrogels also show complex phenotypes, increasing the scope for phenotypic profiling. Here we describe a high-content screening platform that uses invasive human prostate cancer cells cultured in 3D in standard 384-well assay plates to study the activity of potential therapeutic small molecules and antibody biologics. Image analysis tools were developed to process 3D image data to measure over 800 phenotypic parameters. Multiparametric analysis was used to evaluate the effect of compounds on tissue morphology. We applied this screening platform to measure the activity and selectivity of inhibitors of the c-Met and epidermal growth factor (EGF) receptor (EGFR) tyrosine kinases in 3D cultured prostate carcinoma cells. c-Met and EGFR activity was quantified based on the phenotypic profiles induced by their respective ligands, hepatocyte growth factor and EGF. The screening method was applied to a novel collection of 80 putative inhibitors of c-Met and EGFR. Compounds were identified that induced phenotypic profiles indicative of selective inhibition of c-Met, EGFR, or bispecific inhibition of both targets. In conclusion, we describe a fully scalable high-content screening platform that uses phenotypic profiling to discriminate selective and nonselective (off-target) inhibitors in a physiologically relevant 3D cell culture setting.
Article
Full-text available
Improving the ability to predict the efficacy and toxicity of drug candidates earlier in the drug discovery process will speed up the introduction of new drugs into clinics. 3D in vitro systems have significantly advanced the drug screening process as 3D tissue models can closely mimic native tissues and, in some cases, the physiological response to drugs. Among various in vitro systems, bioprinting is a highly promising technology possessing several advantages such as tailored microarchitecture, high-throughput capability, coculture ability, and low risk of cross-contamination. In this opinion article, we discuss the currently available tissue models in pharmaceutics along with their limitations and highlight the possibilities of bioprinting physiologically relevant tissue models, which hold great potential in drug testing, high-throughput screening, and disease modeling.
Article
Full-text available
Liver biology and function, drug-induced liver injury (DILI) and liver diseases are difficult to study using current in vitro models such as primary human hepatocyte (PHH) monolayer cultures, as their rapid de-differentiation restricts their usefulness substantially. Thus, we have developed and extensively characterized an easily scalable 3D PHH spheroid system in chemically-defined, serum-free conditions. Using whole proteome analyses, we found that PHH spheroids cultured this way were similar to the liver in vivo and even retained their inter-individual variability. Furthermore, PHH spheroids remained phenotypically stable and retained morphology, viability, and hepatocyte-specific functions for culture periods of at least 5 weeks. We show that under chronic exposure, the sensitivity of the hepatocytes drastically increased and toxicity of a set of hepatotoxins was detected at clinically relevant concentrations. An interesting example was the chronic toxicity of fialuridine for which hepatotoxicity was mimicked after repeated-dosing in the PHH spheroid model, not possible to detect using previous in vitro systems. Additionally, we provide proof-of-principle that PHH spheroids can reflect liver pathologies such as cholestasis, steatosis and viral hepatitis. Combined, our results demonstrate that the PHH spheroid system presented here constitutes a versatile and promising in vitro system to study liver function, liver diseases, drug targets and long-term DILI.
Article
Full-text available
Background Targeted anti-HER2 therapy has greatly improved the prognosis for many breast cancer patients. However, treatment for HER2 negative disease is currently still selected from a multitude of untargeted chemotherapeutic treatment options. A predictive test was developed using patient-derived spheroids to identify the most effective therapy for patients with HER2 negative breast cancer of all stages, for clinically relevant subgroups, as well as individual patients. Methods Tumor samples from 120 HER2 negative patients obtained through biopsy or surgical excision were tested in the breast cancer spheroid model using scaffold-free cell culture. Similarly, spheroids were also generated from established HER2 negative breast cancer cell lines T-47D, MCF7, HCC1143, and HCC1937 to compare treatment efficacy of heterogeneous cell populations from patient tumor tissue with homogeneous cell lines. Spheroids were treated in vitro with guideline-recommended compounds. Treatment mediated impact on cell survival was subsequently quantified using an ATP assay. Results Differences were observed in the metabolic activity of the untreated spheroids, whereby cell lines consistently achieved higher values compared to tissue spheroids (p < 0.001). A higher number of cells per spheroid correlated with a higher basal metabolic activity in tissue-derived spheroids (p < 0.01), while the opposite was observed for cell line spheroids (p < 0.01). Recurrent tumors showed a higher mean vitality (p < 0.01) compared to primary tumors. Except for taxanes, treatment efficacy for most tested compounds differed significantly between breast cancer tissue spheroids and breast cancer cell lines. Overall a high variability in treatment response in vitro was seen in the tissue spheroids regardless of the tested substances. A greater response to anthracycline/docetaxel was observed for hormone receptor negative samples (p < 0.01). A higher response to 5-FU (p < 0.01) and anthracycline (p < 0.05) was seen in high grade tumors. Smaller tumor size and negative lymph node status were both associated with a higher treatment efficacy to anthracycline treatment combined with 5-FU (cT1/2 vs cT3/4, p = 0.035, cN+ vs cN−, p < 0.05). Conclusions The tissue spheroid model reflects current guideline treatment recommendations for HER2 negative breast cancer, whereas tested cell lines did not. This model represents a unique diagnostic method to select the most effective therapy out of several equivalent treatment options.
Article
Full-text available
The field of regenerative medicine has progressed tremendously over the past few decades in its ability to fabricate functional tissue substitutes. Conventional approaches based on scaffolding and microengineering are limited in their capacity of producing tissue constructs with precise biomimetic properties. Three-dimensional (3D) bioprinting technology, on the other hand, promises to bridge the divergence between artificially engineered tissue constructs and native tissues. In a sense, 3D bioprinting offers unprecedented versatility to co-deliver cells and biomaterials with precise control over their compositions, spatial distributions, and architectural accuracy, therefore achieving detailed or even personalized recapitulation of the fine shape, structure, and architecture of target tissues and organs. Here we briefly describe recent progresses of 3D bioprinting technology and associated bioinks suitable for the printing process. We then focus on the applications of this technology in fabrication of biomimetic constructs of several representative tissues and organs, including blood vessel, heart, liver, and cartilage. We finally conclude with future challenges in 3D bioprinting as well as potential solutions for further development.
Article
Full-text available
To investigate the underlying causes of chemoresistance in malignant pleural mesothelioma, we have studied mesothelioma cell lines as 3D spheroids, which acquire increased chemoresistance compared to 2D monolayers. We asked whether the gene expression of 3D spheroids would reveal mechanisms of resistance. To address this, we measured gene expression of three mesothelioma cell lines, M28, REN and VAMT, grown as 2D monolayers and 3D spheroids. A total of 209 genes were differentially expressed in common by the three cell lines in 3D (138 upregulated and 71 downregulated), although a clear resistance pathway was not apparent. We then compared the list of 3D genes with two publicly available datasets of gene expression of 56 pleural mesotheliomas compared to normal tissues. Interestingly, only three genes were increased in both 3D spheroids and human tumors: argininosuccinate synthase 1 (ASS1), annexin A4 (ANXA4) and major vault protein (MVP); of these, ASS1 was the only consistently upregulated of the three genes by qRT-PCR. To measure ASS1 protein expression, we stained 2 sets of tissue microarrays (TMA): one with 88 pleural mesothelioma samples and the other with additional 88 pleural mesotheliomas paired with matched normal tissues. Of the 176 tumors represented on the two TMAs, ASS1 was expressed in 87 (50%; staining greater than 1 up to 3+). For the paired samples, ASS1 expression in mesothelioma was significantly greater than in the normal tissues. Reduction of ASS1 expression by siRNA significantly sensitized mesothelioma spheroids to the pro-apoptotic effects of bortezomib and of cisplatin plus pemetrexed. Although mesothelioma is considered by many to be an ASS1-deficient tumor, our results show that ASS1 is elevated at the mRNA and protein levels in mesothelioma 3D spheroids and in human pleural mesotheliomas. We also have uncovered a survival role for ASS1, which may be amenable to targeting to undermine mesothelioma multicellular resistance.
Article
Smoking represents a major risk factor for chronic obstructive pulmonary disease (COPD), but it is difficult to characterize smoke-induced injury responses under physiological breathing conditions in humans due to patient-to-patient variability. Here, we show that a small airway-on-a-chip device lined by living human bronchiolar epithelium from normal or COPD patients can be connected to an instrument that “breathes” whole cigarette smoke in and out of the chips to study smoke-induced pathophysiology in vitro. This technology enables true matched comparisons of biological responses by culturing cells from the same individual with or without smoke exposure. These studies led to identification of ciliary micropathologies, COPD-specific molecular signatures, and epithelial responses to smoke generated by electronic cigarettes. The smoking airway-on-a-chip represents a tool to study normal and disease-specific responses of the human lung to inhaled smoke across molecular, cellular and tissue-level responses in an organ-relevant context.
Article
In the Methods of this Letter, '5,000' should have read '15,000' in the sentence: "Then, cells were again plated on a Matrigel-coated at 15,000 cells per cm² in MEF-CM." This error has been corrected online. © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.