3D Models of The Bone Marrow in Health and Disease, Yesterday, Today, and Tomorrow. 2018

You might also like

You are on page 1of 16

MRS Communications (2018), 1 of 16

© Materials Research Society, 2018


doi:10.1557/mrc.2018.203

Prospective Article

3D models of the bone marrow in health and disease: yesterday, today,


and tomorrow

Annamarija Raic, Toufik Naolou, Anna Mohra, Chandralekha Chatterjee, and Cornelia Lee-Thedieck, Karlsruhe Institute of
Technology (KIT), Institute of Functional Interfaces, 76344 Eggenstein-Leopoldshafen, Germany
Address all correspondence to Cornelia Lee-Thedieck at cornelia.lee-thedieck@kit.edu

(Received 3 August 2018; accepted 10 September 2018)

Abstract
The complex interaction between hematopoietic stem cells (HSCs) and their microenvironment in the human bone marrow ensures a life-long
blood production by balancing stem cell maintenance and differentiation. This so-called HSC niche can be disturbed by malignant diseases.
Investigating their consequences on hematopoiesis requires a deep understanding of how the niches function in health and disease. To facil-
itate this, biomimetic models of the bone marrow are needed to analyze HSC maintenance and hematopoiesis under steady state and diseased
conditions. Here, 3D bone marrow models, their fabrication methods (including 3D bioprinting), and implementations recapturing bone
marrow functions in health and diseases are presented.

Introduction with enriched mesenchymal stromal cells (MSCs) which are sub-
The red bone marrow, located in the cavities of the trabecular divided into Leptin+, C-X-C motif chemokine 12 (CXCL12)
regions in long bones, represents a complex blood and bone abundant reticular (CAR) or Nestin+ cells. These cell populations
cell-producing organ. Blood cells develop from immature are an important source of factors required for HSC mainte-
hematopoietic stem and progenitor cells (HSPCs) and ensure nance.[1,5] Apart from these, hematopoietic cells which are
the completion of several essential tasks like immune function, found throughout the bone marrow niches, are also able to influ-
blood clotting or oxygen transport. Whereas most mature blood ence HSC fate.[1] Among others, T-cells in the endosteal region
cells undertake their tasks mainly outside of the bone marrow, regulate stem cell maintenance, whereas an ablation of megaka-
the only place where hematopoietic stem cells (HSCs) execute ryocytes, the early precursor of blood clotting thrombocytes,
their functions (stem cell maintenance and blood cell produc- from the bone marrow results in mobilization of HSCs into the
tion by differentiation) to guarantee lifelong blood production, peripheral blood system.[8,9] The named endothelial, perivascular
is in their stem cell niche in the red part of the bone marrow.[1] or hematopoietic cells are just a selection out of many niche cells,
Current studies revealed the existence of such niches at certain which are all known to regulate HSC function and are important
sites of the bone marrow. These niches are illustrated in a sim- regulators in the environment of HSCs. The niche cells govern
plified form in Fig. 1. One of those is described as the endosteal HSCs by providing cell–cell contacts or by the production of
niche, characterized by bone regenerating osteoblasts (OBs) soluble factors. The most popular cell–cell contacts in the con-
located at the endosteal surface of bone.[1] OBs are able to reg- text of the HSC niche are cell-cell adhesions via N-cadherin,
ulate HSC function e.g., by activation of the Notch pathway interactions, via vascular cell adhesion molecule 1 (VCAM1)
and production of thrombopoietin (TPO), they are involved and very late antigen-4 (VLA-4; integrin α4β1) or the Notch
in controlling HSCs quiescence.[2,3] Another site for HSC ligand–receptor interplay.[10,11] In addition to these, soluble
niches was described close to the vascular structure near the factors such as fibroblast growth factor 2, WNT ligands, stem
endosteum, specified as arterioles, which have also an impact cell factor (SCF) or CXCL12 expressed by MSCs as well as
on HSC quiescence and thus form a unique arteriolar bone morphogenic proteins (BMPs) or angiopoietin expressed
niche.[4] Nevertheless, dividing and non-dividing HSCs are by other niche cells have an impact on cell cycle, maintenance
mainly found in the perisinusoidal regions of the bone mar- of HSCs and normal niche function.[1] Furthermore, niche cells
row.[5] Here, HSCs are in close proximity to blood vessel cov- produce extracellular matrix (ECM) components. The ECM,
ering endothelial cells, which affect HSC dormancy and embedding the cells in their niche, consists of proteins such
self-renewal via cell–cell contacts (e.g., via E-selectin). This as collagen IV, glycoproteins such as fibronectin or glycosami-
region is generally referred to as the endothelial/perisinusoidal noglycans (GAGs, e.g. hyaluronic acid (HA)) and matrix
niche.[6,7] At this perivascular site, HSCs are in close contact remodeling enzymes. This protective shelter functions as a

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪1
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Figure 1. Schematic drawing of the bone marrow niche in vivo. The simplified representation of the niche in the bone marrow shows the microenvironment of
HSCs (light red) consisting of several types of niche cells such as the OBs (blue) located at the endosteum, perivascular endothelial cells (red) or MSCs (purple)
and biochemical stimuli due to the expression of soluble factors (red and yellow dots) by niche cells. Furthermore, biophysical parameters (different stiffnesses
reviewed in Nelson & Roy[18]) throughout the bone marrow which have an impact on cells behavior are illustrated in green.

growth factor reservoir and determines the balance between cues regulate HSC fate and how these signal transductions
stemness and differentiation with the help of cell–matrix inter- are changed in a diseased bone marrow, are required.
actions.[12–15] HSCs can bind to ECM proteins mainly via In order to cope with these scientific tasks, bone marrow
membrane-bound integrins.[16] On one hand, this interaction model systems are needed which (i) allow analysis of human
can result in a protein sequence-specific cell response; on the hematopoiesis in steady state and disease, (ii) can be used as
other hand, the cells can sense the biophysical properties of drug-testing systems and (iii) facilitate the in vitro expansion
the substrate leading to a substrate stiffness-dependent cell of HSCs. For this purpose, animal models are often used,
response. In the latter case, HSCs rearrange their cytoskeleton accepting the major drawback of insufficient transferability to
after substrate binding and exert traction to the substrate. In a human beings.[22] Furthermore, the ethical aspects and in this
process called mechanotransduction, this mechanical stimulus context the implementation of the 3Rs principle to replace,
can be converted into a biochemical signal in the cell, which reduce and refine animal experiments urges alternatives.[23]
can influence HSC behavior.[17] The stiffness in the bone mar- Therefore, in vitro models have steadily been developed during
row affecting HSC behavior ranges from 0.1 to 1 kPa in the the last years. The usage of conventional 2D cell culture
central marrow and 5–20 kPa in the perivascular region up to devices is becoming less common due to the increasing know-
>35 kPa in the endosteal part.[18] ledge of the in vivo conditions revealing several drawbacks of
This summarized overview of bone marrow components the highly artificial 2D environment. The complex interplay of
exhibits that HSCs are embedded in an intricate environment cells with their surrounding in a 3D architecture affects the
in which they receive cell-specific, biochemical, and biophysi- morphology of stem cells as well as their differentiation into
cal signals from all three dimensions. This unique environment mature cells disclosing the importance of a 3D environment
of the bone marrow represents the only place where HSCs are for HSCs.[17,24] In the following chapters, we review
able to maintain their stem cell properties life-long. So far, in state-of-the-art 3D cell culture models mimicking the bone
vitro culturing of HSCs is not sufficient to maintain stem cell marrow in vivo in health and disease as well as methods for
properties and ends up with differentiation of the stem cells their construction with special emphasis on 3D bioprinting.
into committed progenitors or mature blood cells. Sufficiently Furthermore, their implementation for in vitro cell analysis
in vitro multiplied HSCs from one donor could be used for and future aspects in context to refine bone marrow models
cell transplantations into several patients with leukemia and are discussed.
thereby overcome the problem of donor limitations.[19]
Furthermore, malignant diseases concerning the bone marrow Evolution of bone marrow models
can severely alter or even destruct the complex interaction in As already mentioned, improvements of the HSC expansion for
the HSC niche.[20,21] The treatment of such disorders comprises clinical needs and of bone marrow models for fundamental
the medication with disease-specialized drugs.[20] For effective studies and drug-testing are indispensable in future research.
targeting of the disease and to improve the therapeutic out- To reach this goal, HSC cultures which mimic relevant charac-
come, new drugs have to be developed. To achieve these teristics of their natural healthy niche in the bone marrow, need
goals, more information on how biochemical and biophysical to be developed. The evolution of such bone marrow models,

2▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

which comprised more and more aspects of a natural niche and were slightly improved in patients with previous in vitro HSPC
increased in complexity in the last decades, is described in the expansion before transplantations compared with direct trans-
following and summarized in Fig. 2. Conventionally, suspen- plantations without HSPC expansion. However, no significant
sion culture systems are used to expand umbilical cord blood improvement in the survival of the patients was reported, as
HSPCs in research or in clinical studies before transplantations. recently reviewed.[26] Conventional protocols supported the
By adding cytokines to the culture medium, which is drafted in proliferation of HSCs but they did not foster the self-renewal
Fig. 2(I), HSC expansion could be improved significantly. of HSCs to a large extent, which led to a rapid decline of stem-
Commonly used cytokines for HSC expansion are for example ness in such in vitro cultures.[27] With the purpose to improve
TPO, FMS-like tyrosine kinase 3 ligand (Flt3L), SCF, granulo- the expansion of HSCs while maintaining stemness, research-
cyte colony-stimulating factor (GCSF), interleukin-6 (IL-6) ers looked more closely at the natural stem cell niche and
and interleukin-3 (IL-3). Furthermore, several developmental started trials to mimic it in its entirety, including chemical con-
regulators such as the Notch ligand Delta-1 or small molecules ditions, cell and ECM compositions as well as biophysical
such as the copper chelator tetraethylenepentamine (TEPA), properties.
nicotinamide and StemReginin-1 were used in addition to the In this attempt, researchers tried to imitate the ECM of the
cytokine cocktails, which further improved the total cell and bone marrow in many different ways. In standard cell culture,
CD34+ HSPC expansion.[19] Likewise, several cell types that conventional tissue culture plates, which are made from poly-
occur naturally in the bone marrow were described to assist styrene and are thus stiff (E = 3000 MPa[28]), were used to cul-
HSC proliferation and maintenance by secreting HSC-supporting ture HSCs in suspension. By coating the culture devices with
factors in cell cultures, which is delineated in Fig. 2(II). This natural components of the bone marrow ECM, such as fibro-
includes, e.g., OBs, naturally present in the endosteal niche, nectin or collagen, as shown in green in Fig. 2(III), HSCs
endothelial cells, in vivo an important cell type of the vascular were enabled to attach to this matrix. The attachment of
niche, and others that were mentioned before in the intro- HSCs does not only affect the binding itself but also influences
duction. The most commonly used supporting cells in HSC proliferation and stemness of the cell e.g., by mechanotransduc-
cultures are MSCs because they express higher levels of tion, which is why it is important to allow attachment of the
HSC-supporting factors than other stromal cells,[25] and they HSCs also in in vitro cultures. Instead of proteins, also smaller
were already used to expand HSCs for transplantations in clin- bioactive sequences, such as the minimal integrin recognition
ical studies. In clinical studies, neutrophil and platelet recovery motif RGD or the fibronectin fragment CS-1, were applied to

Figure 2. Schematic drawing of the development of 3D models. Evolution of conventional HSPC cultures into sophisticated 3D biomimetic bone marrow niches.
In conventional culture systems, HSPCs are cultivated and expanded on 2D polystyrene devices in combination with cytokines and small molecules (i) or in
addition to feeder cells (ii). By functionalizing the substrate and varying its physical parameters such as stiffness, ECM properties were mimicked in research
applications (iii). Allowing cells to grow in a 3D environment, e.g., as spheroids, embedded in a polymer matrix or in the cavities of pores of polymer scaffolds
(iv), reformed culturing processes of HSPCs. Using bioreactors or microfluidic devices further improved nutrient supply and added more parameters of the niche
to the culture system (v). In future research, more attempts should be made to mimic the niche in more detail, combining various substrates (shown in light
green and light orange), stiffnesses (displayed as brightness of the substrate-colors), cells (depicted by the blue and the purple cell), soluble factors (presented
by yellow and red dots) and a vascular system (shown as a red circle) in one model (vi).

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪3
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
allow the attachment of the cell to synthetic matrices. Placing (except at the spheroid–medium interface) and thus cells can
these proteins and peptides in a controlled manner by nanopat- communicate properly.[37] Currently, a widespread method to
terning added another biophysical parameter to culture systems culture HSCs in 3D is the incorporation of cells into a polymer
since nanoscale topography and nanopatterning also have solution that is cross-linked to form a gel (depicted in green in
effects on cell behavior. For example, scientists found out the second part of Fig. 2(IV)) in the following. The cells are
that a distance less than 45 nm between cyclic RGD peptides encapsulated in the forming hydrogel, which resembles the
is required for proper HSC attachment and that efficient cell- ECM in vivo. Natural or synthetic polymers, which are able
peptide adhesion may foster proliferation and differentiation to swell in water, were used for this purpose. Materials such
of HSCs.[29] Another physical parameter, which influences as fibronectin, collagen, laminin, and GAGs occur in the
stem cell behavior, is the stiffness of the substrate. Cells can bone marrow in vivo and HSCs attach easily to them, which
track the stiffness of a substrate by mechanosensing and there- is why they were often used in 3D cell cultures.[31] Other na-
upon change their behavior. For example, when MSCs were tural materials used for scaffolds were chitosan, alginate,
grown on a stiff substrate, they differentiated and expressed Matrigel™ (gelatinous protein mixture), agarose, and bacterial
markers of osteogenic lineages whereas on soft surfaces they nanocellulose.[38,39] By functionalizing synthetic matrices with
expressed neurogenic lineage markers.[30] For HSPCs it was small peptides, such as RGD, the culture conditions can be con-
found that they adhered better, were more motile and kept a trolled precisely, while native ECM molecules, such as HA, not
higher level of stemness when they were cultured on stiffer only affect adhesion but also influence a lot of other signaling
hydrogels.[31] They proliferated the most if they were cultured pathways.[40] Conventional synthetic polymers used for hydro-
on surfaces with intermediate stiffness (approximately 12.2 gels are, e.g., poly (lactic acid) (PLA), poly (ε-caprolactone)
kPa).[32] (PCL), poly (lactic-co-glycolic acid) (PLGA) and poly (ethyl-
In an in vitro 2D culture it is almost impossible to recipro- ene glycol) (PEG). These materials were used alone or in
cate complex biochemical and biophysical aspects as they conjunction with each other. In some models, inorganic com-
can be generated in 3D in vitro cultures. For instance, the 2D ponents of the bone such as hydroxyapatite (HAP) and trical-
environment does not mimic the 3D bone marrow in vivo in cium phosphate (TCP) were also incorporated.[38,39] Proper
architecture, leading to an irrelevant phenotype of cells.[33] In attachment of cells and cell compatibility are major require-
contrast to a 3D culture, cells in 2D grow in an unnatural dorsal- ments of these gels and the possibility to modulate the gel prop-
ventral polarization affecting their contact surface for cell–cell erties, such as stiffness, is a further advantage to improve them
or cell–matrix interactions, which in turn influences cell mor- as culture systems. For the further usage of HSCs, e.g., in trans-
phology. For example, MSCs flattened when cultured on 2D plantations, it is important that the cells can be efficiently
surfaces, which further affected the tension in the MSC cyto- recovered from the gels without harming them. This require-
skeleton and the expression of pluripotency genes.[34] On top ment could be met by degradable gels. Gels with natural poly-
of that, cells in 2D culture are covered by large volumes of mers can, for example, be degraded by enzymes whereas in
medium. Secreted molecules are diluted immediately in the synthetic materials special cross-linkers (e.g., polymers with
medium and no signal gradients can be created which impairs disulfide bonds), which are cleaved by special stimuli such as
autocrine and paracrine signaling.[35] Due to these drawbacks, light (photocleavable linkers, e.g., Fairbanks et al.[41]) or reduc-
the idea came up to also consider dimensionality in cell cultur- ing agents (e.g., Zhang et al.[42]) can be introduced.[43] Such
ing.[17] Many different 3D cell culture systems with different stimuli-responsive gels gain more and more importance nowa-
complexity were developed and found to support HSC expan- days. An interesting application is the usage of biodegradable
sion and maintenance to a higher level than 2D systems. In the hydrogels. Cytokines, small molecules or other factors that
following part, some of them will be presented. influence cells are incorporated into the gel and undergo a sus-
The simplest 3D in vitro cultures are spheroids. To generate tained release during the process of degradation induced by the
spheroids, cells can be cultured in hanging drops or on non- cells themselves (e.g., Cheng et al.[44]). Also cell migration,
adhesive surfaces to force them to form aggregates. In cell–cell contacts and stemness of the cells was improved, if
Figure 2(IV) a hanging drop culture is sketched, comprising cells were able to remodel their matrix.[45] The advantages of
an aggregate of different cell types within a drop of medium. encapsulation are that cells have full contact with the matrix
An example is the high-throughput coculture system with and that signaling molecules are not diluted quickly.
MSCs and HSCs presented by Cook and colleagues.[36] They However, the cells are often not in direct contact. Since the nat-
used surface modified microwells to support the formation of ural bone marrow does not consist of a homogeneous matrix
microaggregates, which then developed spatial structures. It but is perforated by pores, meshworks made of fibers and
could be observed that, in comparison with 2D cultures, porous sponge-like scaffolds, such as the sketch in the third
MSCs in the aggregates expressed higher levels of hemato- part of Fig. 2(IV), were developed to mimic this architecture.
poietic niche factors and the HSC expansion increased. Such The cells were added after scaffold preparation. Thus, in
spheroids have the advantages that the cells can form proper these approaches, the cells were not directly surrounded by
cell–cell contacts and tissue-like patterns, they keep their natu- the gel, but they were gathered in the pores of the gel. This per-
ral shape, signaling molecules are not diluted into the medium mitted the formation of close cell–cell contacts and a 3D

4▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

architecture for cell-matrix contacts.[17] The pores could be term bioreactor to describe an engineered system that allows
generated by different methods, such as salt leaching, freeze- automated control over medium supply, waste removal, and
drying or 3D printing and by changing parameters of the man- agitation. Bioreactors were combined with 2D or 3D applica-
ufacturing protocols, pore sizes and shapes could be varied. To tions. Rödling et al. developed a bioreactor containing a
mimic the in vivo conditions and to provide an adequate archi- PEGDA-RGD scaffold, which resembles the drawing in
tecture for the 3D organization of the cells, while allowing for Fig. 2(V). It could mimic the niche in an active or in a steady-
enough place for nutrient exchange, pore sizes between 70 and state condition that either supported HSPC differentiation or
300 µm were chosen.[46,47] A full interconnectivity of the pores HSPC maintenance.[35] Microfluidic devices can also accom-
was achieved, so that the medium and the cells could plish a continuous nutrient and oxygen supply and, furthermore,
completely penetrate the scaffold. Examples for successful gradients (e.g., oxygen, calcium ions, cytokines, and small mol-
applications of porous 3D hydrogels for the expansion of ecules) can be generated.[51] Such systems operate with
HSPCs in vitro are the scaffolds of Ferreira et al.[48] and Raic extremely small amounts of fluids (10−6 to 10−15 mL) using
et al.[47]. Ferreira et al. tested several natural polymers as scaf- laminar flow microchannels (1–1000 µm). To mimic blood ves-
folds in combination with either cytokines or MSCs as support sels that are pervading the tissue in vivo, microfluidic devices
and found fibrin in combination with MSCs to be the most were used and in this way, another important parameter of the
promising system with a 3 × 107-fold expansion of CD34+ bone marrow—the mimicry of vasculature and perfusion—
cells compared with conventional 2D systems.[48] Figure 3(a) could be included into models. Kotha et al. developed a sophis-
shows a scanning electron micrograph of HSCs and MSCs ticated model that mimics very well the vascular niche and the
attaching to a synthetic poly (ethylene glycol) diacrylate process of HSC homing in a microfluidic device, which com-
(PEGDA)-RGD scaffold. Raic et al. showed that HSPC expan- prises various cell types, a flow system, and a soft matrix.[49]
sion was upregulated in this scaffold in comparison to the They constructed an endothelialized vascular network in a col-
expansion in 2D tissue culture plates if HSPCs were cocultured lagen matrix with MSCs and other bone marrow stromal cells.
with MSCs.[47] After perfusing the network with medium containing monocytes
To culture large amounts of cells in an active state over a (CD14+/CD45+) or HSPCs (CD34+/CD45+), they could
long period of time, bioreactors were used. Here, we use the observe these cells adhering to or migrating into the soft matrix.

Figure 3. In vitro models of the bone marrow in health and disease. (a) Example of a bone marrow niche model in healthy conditions: Pseudo colored scanning
electron micrograph of a porous PEGDA-RGD hydrogel scaffold (blue) from Raic et al. The 3D culture system was seeded with MSCs (purple) from the bone
marrow and HSPCs (red). Arrows indicate the size of the cells (MSC ≈ 28 µm, HSPC ≈ 12 µm). Scale bar = 20 µm. Adapted/reproduced with permission from
Elsevier, ref. 47 (b) In vitro model of a leukemic niche: Microvessel system lined by endothelial cells mimicking the vascular niche developed by Kotha and
colleagues. The LCs (blue) were perfused in the microvessel system which was embedded in collagen type I (left). Addition of fibroblasts (HS5-cells; green) into
the matrix around the vessels led to increased adherence and migration of LCs (right). Scale bars = 100 µm. Adpated/reproduced with permission from BioMed
Central, ref. 49 (c) A model simulating prostate cancer bone metastasis: On the left is a scanning electron microscopy image of the scaffold prepared from
mPCL-TCP and then seeded with human OBs. Within 3 weeks, OBs formed a sheet on the scaffold. Scale bar = 2 mm. On the right is the immunofluorescent
staining of OBs with phalloidin (red) and PC-3 cells with anti-pan CK (green) after 6 days of coculture. PC-3 cells are characterized by a spherical (white open
arrow) and fibroblast appearance (white closed arrow). Scale bar = 75 µm. Adapted/reproduced with permission from Taylor and Francis Ltd. ref. 50.

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪5
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Very complex microfluidic systems are organs-on-a-chip. environment resembling the in vivo conditions. In the follow-
An interesting example is the bone marrow-on-a-chip from ing part, in vitro models are presented that allow investigations
Torisawa and colleagues.[52] They implanted a collagen gel of these regulatory cell interactions and contribute new hints
with demineralized bone powder and BMP subcutaneously about the function of the human leukemic bone marrow.
into a mouse where it developed into a bone with an intact Similar to HSCs, LCs are also found in the vascular or
bone marrow. Afterward, the bone was taken out and put into endosteal part of the bone marrow and can interact with
a microfluidic device where it could be cultured for at least 1 cell-populations of these microenvironments.[64,65] Bray et al.
week. This system proved to be a proper in vitro model for irra- constructed a 3D PEG-heparin hydrogel with human umbilical
diation toxicity tests. However, it was of murine origin and did vein endothelial cells (HUVECs) amongst others as environ-
thus not represent the human bone marrow. Reinisch et al. devel- mental stimuli to investigate the impact of the vascular niche
oped humanized bone marrow-ossicles by implanting human in acute myeloid leukemia (AML). They found that cells
MSCs into mice and let them form bones, which were then from the vascular network in a 3D scaffold support the resis-
chimeric, of murine and human origin.[53] Such microfluidic sys- tance of LCs against chemotherapeutics.[66] Furthermore,
tems are also one of many possible approaches to mimic the Kotha and colleagues also developed an artificial vascular
bone marrow in malignant diseases by imitating the invasion microenvironment which allowed studying 3D multicellular
of cancer cells from the bloodstream into the bone marrow in interactions and cell trafficking between distinct fibroblasts
vitro. In the following two chapters, such approaches for models and LCs. In the in vitro vascular marrow, they showed that
of leukemia and bone metastasis will be presented and discussed. LCs respond to distinct fibroblasts which were embedded in a
collagen type I matrix around the vessels with increased adhe-
Leukemic niche models sion and migration within the vessel network which is shown in
In hematological disorders such as leukemia, the bone marrow Fig. 3(b).[49] In an engineered microfluidic 3D collagen matrix,
is overflooded by malignant hematopoietic cells leading to a presenting an endosteal in vitro platform, Bruce and colleagues
disturbed or insufficient hematopoiesis.[54] Transformation of showed direct cell–cell interactions in 3D between MSCs and
hematopoietic cells into malignant cells can occur due to in vivo endosteally located OBs with LCs from acute lympho-
genetic events, hereditary or environmental conditions and blastic leukemia.[67] Using in vitro transwell filter devices
affect the myeloid or lymphoid cell lineage.[54,55] A small pro- revealed an influence of MSCs, which represent the main pop-
portion of these cells regain stem cell properties, such as an ulation in bone marrow stroma, on survival and viability of LCs
unlimited self-renewal potential and the ability to initiate leuke- isolated from patients with chronic lymphatic leukemia. In
mia as well as a reduced differentiation potential.[54,56] The addition, they found that a coculture of MSCs with leukemic
malignant leukemic cells (LCs) occupy the bone marrow B cells promote the cytokine production of MSCs, concluding
niche for their own survival and an uncontrolled proliferation that LCs are able to rearrange their cytokine milieu.[68] These
contributing to disease progression.[57,58] The highly organized extracellular communication molecules are important for cell
HSC niche is rearranged by LCs and a leukemic niche is homing, migration and growth. Changing their composition
formed, accompanied by a loss of important signals and by increasing the expression of homing or anti-apoptotic fac-
niche cells, impairing normal homeostasis.[58] In this protective tors in the bone marrow can support LCs survival.[59,69]
environment, LCs are not assailable for some therapeutic pro- Therefore, studying cell behavior in response to soluble mole-
cedures aiming at the destruction of malignant cells.[59] Due cules is of great importance to find new treatment strategies that
to their acquired stem cell properties and the interception of are based on abrogating the connection between LCs and their
anti-apoptotic signals, they can survive in the marrow despite supportive environment. With the objective of finding molecu-
harsh conditions during treatment and can lead to a reappear- lar mechanisms to reach this goal, migration studies in a 3D fil-
ance of the disease.[54,59,60] Development of new therapeutic ter system revealed the involvement of the Raf/MEK/ERK1/2
strategies to address this problem needs in vitro tools overcom- pathway in the cell response of leukemic T cells after binding
ing the limitations of animal models, in most cases mouse- of the CXCL12 homing factor.[70] CXCL12 is known to medi-
based model systems.[61] Apart from the obvious discrepancies ate chemoresistance after binding to its receptor C-X-C chemo-
between human and mice, such as bodyweight, lifespan or fer- kine receptor type 4 (CXCR4) on the cell surface of LCs in
tility, the differences in the assembly of the peritumoral milieu different leukemia types.[71,72] Due to its great importance, sev-
limit the often used xenotransplantation models which allow eral 3D cell culture platforms were developed to analyze the
the investigation of human cells in an in vivo system.[61–63] impact of the CXCL12/CXCR4 axis on LCs migration.
Therefore, in vitro models gain more and more importance in Promising in vitro studies concerning disruption of these inter-
investigating the origin and the progression of hematopoietic actions showed a decreased drug resistance of chronic myeloid
malignancies and the associated signaling pathways to develop leukemia cells after pretreatment with a CXCR4 inhibitor.[73]
new treatment strategies. For this purpose, current studies are The combination of a CXCL12 inhibitor and a tyrosine kinase
focused on the establishment of in vitro models which allow inhibitor reduced cell migration to a greater extent than either
analysis of cell–cell and cell–matrix interactions as well as drug alone.[74] In the endosteal region of the bone marrow,
the interplay between LCs and soluble factors in a 3D OBs express osteopontin which can bind to its receptor on

6▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

LCs leading to protection against apoptosis. In a 3D polysty- into the blood and lymph vessels (intravasation). These circu-
rene scaffold coated with OBs and LCs, the addition of an lating tumor cells translocate to secondary organs, fighting
inhibitor for the osteopontin receptor together with a tyrosine immune resistance and tolerating mechanical forces in the pro-
kinase inhibitor disrupted the adhesion between the LCs and cess. At this point, they exit from the bloodstream (extravasa-
the protective microenvironment and increased their sensitivity tion), settle in and adapt to the new microenvironment. Now,
to drugs. Therefore, blocking the LC-binding to osteopontin the disseminated tumor cells may either enter dormancy as sin-
could be an efficient therapy for patients with AML.[75] Cells gle cells or as small clusters (micrometastases), during which
encapsulated in a 3D surrounding not only communicate with they develop therapeutic resistances and long-term survival
other niche cells and their expressed cytokines but also their strategies. The disseminated tumor cells deploy survival signals
interactions with the ECM allocate cell behavior. Porous scaf- which give rise to drug-resistant clones and finally proceed to
folds fabricated from polymeric materials coated with the ECM form macrometastases.[81,83,84] Events behind metastasis,
protein collagen were proven to be suitable for the culture of such as the invasion of circulating tumor cells, growth and sur-
LC lines in contrast to uncoated controls, highlighting the vival of disseminated tumor cells, evasion of host-tissue
importance of cell-ECM interactions in cell growth.[76] defenses, and amplification of survival signals are not only
Furthermore, migration experiments in a 3D transwell assem- dependent on the innate characteristics of the primary tumor
bly showed an improved migration of a leukemic myeloid but also on the secondary tissue microenvironment. Different
cell line during culture with collagen IV.[77] Investigations of types of cancer show organ-specific metastasis, simultaneously
matrix mechanics and its role on LCs revealed that matrix stiff- or sequentially. Prostate cancer predominantly spreads to the
ness of 3D alginate hydrogels regulates proliferation of AML bone, breast cancer to bone, liver, brain, and lungs and colorec-
cell lines through autocrine signaling. In addition, matrix stiff- tal cancer begins with liver metastasis and moves on to lungs
ness, as well as the integrin ligand (RGD) functionalized scaf- and brain.[85,86]
folds modulated resistance of AML cells against protein kinase About 70% of breast and prostate cancers lead to skeletal
inhibiting drugs.[78] 3D alginate hydrogels, mimicking the soft metastases, followed by thyroid, lungs, kidney and bladder car-
marrow, supported cell growth to a higher extent than the 2D cinomas.[38,82] Multiple myeloma can also be considered as a
control, led to more diverse myeloid progenitor cells and had bone metastatic disease as it not only originates in the bone
an impact on cell cycle.[79] The discussed studies indicate but also spreads throughout the bone marrow.[82,87,88] Bone
that the bone marrow microenvironment including cell–cell, metastasis has an extremely poor prognosis and is mainly char-
and cell–matrix contacts as well as soluble factors play a critical acterized by severe chronic pain, impaired mobility, hypercal-
role in leukemia progression and survival and protects the cemia, spinal cord compression, pathological fractures, and
malignant cells against chemotherapeutic treatment. Results bone marrow aplasia. The appearance of these symptoms sig-
from a previous study, showing that the drug sensitivity of nificantly decreases the quality of life of the patient and, in
LCs depended on the dimensionality of the environment, further most cases, results in death.[38,82,87]
stress the need for new 3D in vitro cell culture platforms.[67] So, what is the reason behind such organ-specific metasta-
Rödling et al. developed a reactor system for perfusion of 3D sis? In 1889, Dr. Stephan Paget, an English surgeon, put for-
scaffolds mimicking the bone marrow in vivo. They could ward his “seed and soil” hypothesis, shedding some light on
demonstrate the relevance of perfusion during drug treatment this phenomenon. He proposed that specific organs provide a
as results differed with and without perfusion.[35] Therefore, comfortable home or the “soil” to certain tumor cells, the
using 3D systems can not only provide new information about “seed”. Progression of metastatic colonization occurs only
the regulation of LCs in an in vivo like environment but is when “the seed” has found its hospitable “soil”.[81] The bone
also a promising tool as a drug-testing platform to find new marrow is a highly vascularized tissue possessing a rich milieu
therapeutic strategies for patients with leukemia. of cytokines, chemokines and growth factors. It also harbors an
arsenal of different cell types such as immune cells, MSCs,
Bone metastasis models HSPCs, bone-forming OBs and bone-resorbing osteoclasts,
Hematological malignancies such as leukemia, lymphoma, and which express specific molecules that facilitate tumor inva-
multiple myeloma are not the only cancers affecting the bone. sion.[89] Kang and colleagues showed that osteolytic factor
Cancer may arise in any organ of the human body. However, it IL-11, connective tissue growth factor, membrane receptor/
is the metastases, rather than the primary tumor, that result in adhesive proteins CXCR4 and osteopontin as well as the metal-
majority of the cancer-related deaths.[80] Metastasis is the mul- loproteinase1 (MMP1) are important parameters that lead
tistage progression of cancer cells from the primary tumor to breast cancer cells to metastasize to bone.[90]
other organs, which, despite extensive research, remains the Stem cell niches present in the bone marrow are of particular
most enigmatic phenomenon in cancer pathophysiology.[80,81] interest to the cancer cells. Shiozawa et al. presented that pros-
In fact, bone occupies the third position among the metastatic tate cancer cells and HSCs actively compete with each other for
sites, briefly trailing behind lungs and liver.[82] the occupation of endosteal niches. Bone marrow stromal cells
Metastasis is a complex series of events beginning with the and OBs express adhesion molecules annexin A2 (AXA2) and
detachment of cancer cells from the primary tumor and entry CXCL12 and facilitate the CXCR4 and the AXA2-expressing

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪7
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
cancer cells and HSCs to bind and home to the bone marrow. angiogenic factors, associated with bone metastasis, were
When the cancer cells succeed in hijacking the niche, they strongly expressed in 3D rather than 2D.[95] Silk has proven
force the HSCs to differentiate terminally.[91] to be a promising material for bone metastatic models owing
Such settings provide the perfect hotbed for the cancer cells to its biocompatibility, strength, toughness, porosity, thermal,
to invade, thrive and expand. The bone always undergoes and chemical stability.[88] Kwon et al. demonstrated the “osteo-
active remodeling which requires a strict balance between mimicry” or bone cell-like behavior of metastatic prostate can-
OBs and osteoclasts. However, during metastatic colonization, cer cell line PC-3 when cultured on silk scaffolds. They
this equilibrium is disrupted and leads to an abnormal increase displayed migration, invasion, survival, and proliferation simi-
in either cell type.[83] Osteolytic lesions are usually common in lar to their in vivo environment.[96] Similar observations were
breast cancer and multiple myeloma whereas prostate cancers made by Cox and colleagues, by conducting experiments
are more prone to osteoblastic metastasis.[82,87] This is why it with breast cancer cells in collagen-GAG scaffolds.[97]
is important to investigate cancer metastasis in a bone marrow Fitzgerald et al. prepared scaffolds using nanoHAP and
microenvironment. collagen and cocultured prostate cancer cells (PC-3 and
In general, tumor microenvironment (TME) is the amalga- LNCaP) with OBs. MMP9, a classic marker for prostate cancer
mation of all cellular and extracellular components surrounding invasiveness, was enhanced in comparison with the 2D coun-
cancer cells with which they maintain an effective crosstalk. terparts.[98] Synthetic polymer-based scaffolds are also reliable
Cancer-associated fibroblasts, transformed epithelial cells, tools for the mechanistic understanding of bone metastasis. In
tumor-infiltrating MSCs, immune cells, adipocytes, and endo- 2010, Sieh et al. fabricated tissue-engineered bone constructs
thelial cells of the blood and lymphatic systems are some of using medical grade PCL and TCP and then seeding them
the cellular constituents of the TME. Cadherins are the most with human OBs isolated from bone tissue. In Figure 3(c) we
important molecules aiding interactions among the cellular can see that after an incubation period of 3 weeks in osteogenic
components whereas cell–matrix interactions are usually estab- medium, a mineralized sheet of OBs was formed and the scaf-
lished by integrins present on the cells. In the TME, various folds were encapsulated in it. Prostate cancer cells (PC-3 and
growth factor- and cytokine-mediated signalings take place, LNCaP) were seeded on these constructs and an augmentation
in an autocrine or a paracrine manner.[92–94] Such interactions of MMP2 and 9 was noted in the cocultures. Prostate-specific
with the tumor cells influence various biologic characteristics antigen, which is also a hallmark of prostate cancer progres-
such as migration, proliferation, quiescence, immune, and ther- sion, was significantly upregulated in the cocultures.
apeutic resistance. The presence of a physicochemical gradient Interestingly, all of these markers were absent in the monocul-
in an avascular tumor mass not only limits the access of oxy- tures without OBs.[50]
gen, nutrients, and soluble factors but also alters the diffusion Cancer cell dormancy is also an important issue that needs
profile of drugs into the tumor.[92,93] Apart from biochemical addressing. Marlow and colleagues devised coculture systems
events, changes in cellular rheology is also a hallmark of cancer involving different metastatic breast cancer cell lines
metastasis. As the formation of macrometastasis progresses, (MDA-MB-231, MDA-MB-453, SUM159, MCF7, BT474,
cells are exposed to a myriad of physical forces including ZR75-1, T47D) with bone marrow-derived MSCs in a 3D col-
hydrostatic pressure, shear stress, compression, and tension lagen biomatrix. The cellular compositions mimicked suppor-
forces.[89] In an in vitro 2D culture it is almost impossible to tive and inhibitory niches. They proved that the cellular
reciprocate a multitude of biochemical and biophysical aspects. components of the TME play a pivotal role both in inducing
Therein lies the importance of in vitro 3D models, which pro- dormancy and in re-entering the cell cycle.[99]
vide researchers with a wider horizon. Cancer cells can be cul- Since bioreactors and microfluidic systems provide an
tured alone or in conjunction with other cell types in a opportunity to implement parameters like vascularization, sev-
spatially-relevant manner, supporting cell–cell and cell–matrix eral research groups have used them to establish the bone mar-
interactions.[38,89,93] A lion’s share of such studies has incorpo- row microenvironment and recreate naturally occurring forces
rated breast and prostate cancer cells. Apart from them, myeloma, in the TME.[38,94] Dhurjati and coworkers have used a bioreac-
lung, renal, colorectal and ovarian cancer cells have also been tor to recapitulate cancer–stromal cell interactions in a bone
investigated in mono or multi-cultures with TME components.[38] marrow microenvironment by coculturing osteoblastic tissue
Naturally-derived materials, which are commonly utilized in with the highly invasive breast cancer cell line MDA-
hydrogel fabrication, influence cancer cell adhesion, and their MB-231. A 3D osteoblastic tissue was grown from isolated
metastatic potential. HA is a preferred substance for bone mar- pre-OBs for up to 5 months in a specialized bioreactor during
row scaffold formations due to its mechanical properties.[88] which active bone formation was observed in the form of
Pan et al. performed a comparative study between 2D and 3D OBs maturation to osteocytes. In the coculture, the cancer
culture systems using the renal carcinoma cell line 786-O in cells penetrated the mature osteoblastic tissue (lower
thiol-modified HA and PEG-based scaffolds. Compared to cell-ECM ratio and mainly osteocyte phenotype) by ECM deg-
the 2D cultures, there was a strong resemblance between the radation. Downregulation of collagen and osteocalcin synthesis
gene expression patterns of the cells in 3D culture to those and upregulation of pro-inflammatory IL-6 cytokine proved
observed in vivo. Adhesion molecules, osteolytic and disruption of osteoblastic bone formation.[100] In further studies

8▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

by Krishnan et al. osteoclasts were incorporated in the cocul- Producing such types of sophisticated scaffolds and models
ture, effectively recreating the active bone remodeling process. is, in particular, imperative in the field of bone tissue engineer-
Degradation of collagen-rich matrix confirmed active osteoclast ing in order to: (i) fabricate vascularized artificial grafts, which
resorption and the addition of the breast cancer cells further can easily integrate in the human body, for the treatment of jaw
aggravated the process. They underwent chemotaxis towards bone defects as well as zygomatic bone fractures,[106,107] (ii)
the active remodeling site and formed colonies comprising build in vitro models to develop and test new treatment strate-
osteoclasts, cancer cells, and pre-osteoclasts. This tri-culture gies for bone disease, (iii) get a deeper understanding of the
model could effectively mimic the “vicious cycle” of bone emergence and evolution of various types of hematological
metastasis and can be considered an interesting tool to evaluate and bone-related diseases and (iv) be used as a platform for
the efficiency of therapeutic drugs in skeletal metastasis.[101] expanding HSCs for transplantation. The term “3D bioprint-
By exercising meticulous spatial and temporal control over ing” is usually used to describe the process of patterning and
the gradients of soluble factors and cell–cell contacts, micro- assembling living and non-living materials to produce bioengi-
fluidic systems can be used to replicate the physiological sig- neered structures.[108] In the following, we use the term 3D
nals in a TME and hence be used to observe tumor invasion, printing to describe the printing of non-living materials and
progression, and neovascularization.[94] A 3D microfluidic 3D bioprinting if living components such as cells are used.
model developed by Bersini et al. recreated a vascularized Almost all known 3D printing techniques have been used to
bone microenvironment with a triple culture using MDA- prepare 3D bone scaffolds. including inkjet bioprinting,[109]
MB-231 cells, osteogenic-differentiated human bone marrow- laser-assisted bioprinting,[110] laser-aided gelling,[111] 3D print-
derived MSCs and HUVECs. It was revealed that extravasation ing with a heatable nozzle,[112] fused deposition modeling
was significantly higher in MSC-conditioned medium and (FDM),[113] extrusion,[114] stereolithography (SLA),[115] and
within 5 days the extravasated cells proliferated to form micro- laser sintering.[116] Table I shows a comparison between differ-
metastases ranging between 4 and 60 cells. Bone-secreted ent printing/bioprinting techniques that have been used in this
C-X-C motif chemokine 5 and breast cancer cell C-X-C field.
motif chemokine receptor 2 played important roles in the Broad spectra of natural and/or synthetic biomaterials in form
extravasation process.[102] of either bioceramics, composites, i.e., a mixture of bioceramics
In the recent past, scores of works have been published and polymers, or polymers have been used as bioinks for 3D
which integrate combinations of cancer and supportive cell printing of bone tissue.[120] This wide diversity of biomaterials
types in a variety of 3D in vitro bone marrow models. and composites that have been used in the 3D printing of bone
Indeed, these models have unfolded several avenues in the scaffolds or grafts allowed scientists to tune the final biomechan-
complex molecular mechanisms involved in the bone metasta- ical and biophysical properties of the constructed scaffolds or
sis and helped in making headway in therapeutic studies. They grafts beside their biodegradability, biocompatibility, and osteo-
have also averted the time and resource-consuming in vivo genic ability that allow differentiation of MSCs to OBs.
models and unsuccessful clinical trials. However, an efficient, Nevertheless, in vast majority of these scientific efforts, a
reliable and cost-effective tool is yet to be commercially simple approach to mimic the bone tissue has been used
available. which was based on 3D bioprinting of several layers with well-
defined thickness. Each of these layers was composed of paral-
3D printing of bone and bone marrow lel lines that were vertically aligned to the lines of the next and
tissues previous layers to yield interconnected porous scaffolds with
3D printing emerged as an innovative solution to address many pore diameters that were mainly determined through the spaces
technical challenges that scientists have been facing in various between the printed parallel lines of each layer. Presence of
fields of science, engineering, and medicine.[103] This tech- interconnected pores is critical for cell survival as it allows
nique is promising in the field of biofabrication as it allows pre- the diffusion of nutrients as well as oxygen into the printed
cise control over placement of different cell types and scaffolds and could allow the vascularization process.[121]
biomaterials.[104] Therefore, 3D printing has attracted scientists These simple models, however, could not fulfill some essential
who are working in the field of tissue engineering as it could requirements that are needed to develop reliable bone or bone
facilitate the preparation of novel 3D scaffolds which could marrow models as well as bone grafts that could mimic the hier-
recapitulate the hierarchical complexity of different types of archical, biophysical and biologic complexity of native bone
biologic tissues, which is not achievable by following the con- tissue. This is mainly attributed to some technical hurdles of
ventional methods of 3D scaffold preparation. Additionally, 3D current 3D bioprinting techniques, as they do not allow simple,
printing potentially allows not only to construct hierarchical 3D simultaneous and regional deposition of several different bio-
hybrid scaffolds with specific physical and biologic character- materials, composites, growth factors, stem-, progenitor- and
istics using heterogeneous materials and biologic cells but also differentiated cells with high resolution and within an accept-
to personalize the final morphological shape based on a able printing speed.[122,123] Some elegant strategies, however,
scanned image obtained using computed tomography (CT) of have been made recently in this direction and a few of them
a patient which would revolutionize treatment processes.[105] are highlighted here. Atala and his research team developed a

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪9
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
https://doi.org/10.1557/mrc.2018.203
Downloaded from https://www.cambridge.org/core.
MRS COMMUNICATIONS • www.mrs.org/mrc 10 ▪

Table I. Comparison of 3D printing/bioprinting techniques.


University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.

Technique Types Background Advantages Disadvantages References

Inkjet bioprinting • Thermal Based on ejecting droplets of liquid • High printing speed (1–10 • Nozzle clogging when bioinks contain [109,117,118]
• Acoustic bioinks either by application of 000 droplets per second) fibers or high cell densities
electrical heat or by application of • High cell viability (>85%) • Non-uniformity of droplet size
voltage to piezoelectric crystals • Low costs • Possible cross contamination when
• Low preparation time printing multiple bioinks
• Biological compatibility simultaneously
with wide array of • Limited to liquid bioinks
biological materials
• Allow potentially printing
of concentration gradients
of cells, materials or
growth factors

Microextrusion • Heatable nozzle Based on extrusion of moderately to • Compatible with a wide • Low printing resolution hinders [112–114,119]
printing/ • Low temperature extrusion highly viscous biomaterials or range of biomaterials’ fabrication of constructs with
bioprinting using pneumatic or bioinks through a tiny nozzle to viscosities vascular microchannels
mechanical (piston or screw) form filaments • Facilitates bioprinting of • Intermediate cell viability due to cell
dispensing systems vascularized constructs damage caused by shear stress when
• Low cost using highly viscous bioinks, small
• Low to medium nozzle diameters and large
preparation time dispensing pressure
• Allow bioprinting of
scaffold-free bioinks

Laser-based • Laser-assisted bioprinting Focusing pulses of laser beam onto • High cell viability due to • High costs [110,111,115]
printing/ (LA) an absorbing substrate to generate absence of nozzle • Long preparation time
bioprinting • Selective laser sintering (SLS) pressure that ejects the bioink • High printing resolution • Inability to 3D print heterocellular
• Laser-aided gelling layer (LA) or onto a thin layer of • Facilitated construction of constructs
biomaterial (SLS) vascularized scaffold • Limited commercial availability
Prospective Article

multi-dispensing printing module, named “integrated constructed scaffold were fabricated using biodegradable
tissue-organ printer” (ITOP), which allowed simultaneous PLA fibers printed with an FDM printer, which resulted in
printing of various cells and polymers through a sophisticated cylindrically shaped constructs containing a series of intercon-
printing nozzle with high printing resolution.[124] The ITOP nected vertical and horizontal vascular channels. This was fol-
system was equipped with multiple cartridges to facilitate the lowed by coating the surface of the construct with dopamine as
printing of multiple cell-laden composite hydrogels beside a prelude to immobilize BMP2, to facilitate the osteogenic dif-
the supporting polymer (PCL) and sacrificial pluronic F-127 ferentiation, via mussel-inspired chemistry. Afterward, human
hydrogel. This allowed the formation of microchannels in the MSCs were seeded into the scaffold. Next, the elastic vascular
scaffold structure which could facilitate the diffusion of nutri- structure was bioprinted using SLA printer, which could fill the
ents and oxygen into the printed tissue as it is shown in interconnected channels with gelatin methacrylate (GelMA)
Fig. 4(a). A human-scale mandible bone construct, seeded hydrogels containing human MSCs and HUVECs. Vascular
with human amniotic fluid-derived stem cells, could be fabri- endothelial growth factor peptide was attached to the GelMA
cated, using ITOP technique, with a geometric structure during the printing process via “thiol-ene” click reaction.
designed based on the CT scan of a human mandible defect Interestingly, the constructed scaffold showed disparate
after traumatic injury which can be seen in Fig. 4(b). In fact, mechanical properties based on region, the compressive modu-
such combination of ITOP technique and CT scan of bone tis- lus of about 0.38 GPa in the stiff parts of the construct whereas
sue could open the door towards reliable models of bone mar- in the vascular region the elastic modulus was in the range of 10
row. Cui et al. presented an integrated approach to fabricate a to 30 kPa, which mimics native bone tissue. This strategy of
hierarchically-vascularized bone construct with a biphasic pattering two adjacent regions with different mechanical and
structure by utilization of a dual 3D printing/bioprinting system biologic properties and coating with different growth factors
composed of FDM and SLA printing techniques.[121] The fab- could support one day the current efforts to fabricate a reliable
ricated construct had a geometric structure and biologic func- bone marrow model since, as it was mentioned in the introduc-
tion mimicking the osteon or Haversian system of natural tion, a native bone marrow owns several niches. Another
bone as it is depicted in Fig. 4(c). The stiff regions of the advanced technological approach that might be used also in

Figure 4. Schematic drawings of some advanced 3D printing techniques for biofabrication. (a) Illustration of the basic 3D architecture which could be achieved
by using the ITOP technique to print multiple-cell hydrogels and supporting PCL polymers. (b) The upper left represents a mandible bone reconstruction
designed based on a human CT image of a mandible bony defect, the upper right image shows a photograph of the 3D printed construct after culturing it in
osteogenic medium for 28 days, and the lower image depicts osteogenic differentiation of stem cells in the printed construct confirmed by Alizarin Red S
staining. Adapted/reproduced with permission from Springer Nature, ref. 124 (c) Schematic illustration of designing and 3D printing of a biomimetic
vascularized bone scaffold, the upper image shows the computer modeling of vascularized bone, the middle images depicts the FDM/SLA 3D bioprinting
platform, while the lower image represents the microstructural design of vascularized construct. Adapted/reproduced with permission from John Wiley and
Sons, ref. 121 (d) Schematics illustrating the design of 3D printer with seven-channel printhead which is in turn connected to seven cartridges that are
individually regulated by programmable pneumatic valves. (e) Side view of one end of a printed microfiber showing its internal 3D structure consisting of seven
individually segmented bioinks. Adapted/reproduced with permission from John Wiley and Sons, ref. 125.

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪ 11
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
the future to support the scientific efforts in this field was the occurrence of bone malignant diseases. Therefore, develop-
recently reported by Khademhosseini and his research team ing a reliable model that can precisely mimic the different niches,
who developed an extrusion 3D printer, which is shown in appears to be mandatory [Fig. 2 (b, VI)]. In order to reach this
Figs. 4(d) and 4(e) that was able to achieve fast and continuous goal, several interrelated parameters which should be taken
extrusion of several materials, i.e., bioinks, with different prop- into consideration are: (i) chemical and biochemical conditions,
erties through one nozzle.[125] e.g., gradient concentration of calcium ions and sustained release
Despite the great potential of 3D printing in the field of of growth factors, (ii) cellular composition, (iii) binding sites for
biofabrication, this technique has not been used yet in the cell attachment, (iv) nanotopography and –patterning, (v) stiff-
development of a reliable model of bone marrow tissue that ness of the different niches, (vi) 3D architecture, (vii) tailorable
recapitulates the exact anatomy and physiology of the different degradation rates, (viii) suitable supply of nutrient and oxygen,
niches in bone marrow, but rather to construct simple models of and (ix) vascularization of the system. Reaching this aim will
porous scaffolds with well-controlled pore size and pattern. enable scientists, for instance, to construct reliable leukemic
Zhou et al., for example, used a tabletop SLA 3D printer to fab- models which are able to support the diffusion of soluble factors,
ricate several 3D highly porous and interconnected scaffolds resembling the in vivo gradient. It should allow also cell migra-
with different geometric patterns.[126] PEGDA containing tion along the gradient and provide an architecture with enough
RGD and/or nanocrystalline HAP were used as bioinks to fab- space to analyze cell–cell contacts in in vitro cocultures. Such a
ricate these scaffolds. The authors investigated the effects of constructed model facilitates the investigation of the anchorage
pore sizes, geometric patterning, and presence of low intensity of LCs in the leukemic bone morrow in consequence of other
pulsed ultrasound on the adhesion, the growth and osteogenic cells or soluble factors in their environment. A large number
differentiation of MSCs. Braham et al. fabricated an advanced of studies has been dedicated to the biochemical pathways
model of 3D bone marrow in order to study the expansion and associated with metastatic progression, but the associated
interaction of primary myeloma cells.[127] The model was com- biophysical cues have been highly underrepresented. These
posed of two separate but interacting niches, i.e., the endosteal parameters are equally important in tumor pathophysiology
niche, prepared by using a bioprintable paste of calcium phos- and investigating them could open doors for new therapeutic
phate cement with seeded osteogenic multipotent MSCs and strategies.
the perivascular niche, which was prepared using Matrigel™ Besides enabling fundamental research in the context of
containing endothelial progenitor cells and MSCs. This was human healthy and diseased bone marrow, bone marrow mod-
the first time that both—the endosteal and perivascular compo- els undeniably serve an array of applications, such as in vitro
nents of HSC niches—were included in one mold, of which one HSC expansion for therapeutic treatment of hematological
part, the endosteal one, was 3D printed. malignancies and as tools for drug-testing. Preparing such
However, as it was mentioned above, even by 3D printing, models using 3D culture techniques, has opened up new vistas
obtaining an ideal bone marrow model is not straightforward, to replicate the complexities present in an in vivo microenviron-
but rather faces several critical technical challenges regarding ment. As discussed earlier, these systems exert a significant
the currently available 3D printing techniques as well as the uti- advantage not only over conventional 2D culture systems but
lized bioinks. Regarding the technical aspect, developments also on in vivo models. They facilitate co-localization of differ-
should be made in order to increase the speed of printing and ent cell types, equip perfusion of biochemical factors and pro-
to facilitate precise and simultaneous bioprinting of several vide a mechanically stable ECM in 3D, all the while
types of bioinks in a way that they would not mix with each circumventing the investment of resource and time necessary
other during or after the printing process and does not cause for in vivo studies. They have also imparted valuable insights
damage to the printed cells. In order to achieve this goal, the into the multifaceted nature of cancer biology and aided studies
development of 3D bioprinters which are able to use two or on leukemic niches and tumor dissemination to the bone. Over
more printing techniques within one printer, and thereby facil- the past years, numerous bone marrow models have been
itating printing of several biomaterials of different nature using researched and several strategies have been developed.
one 3D printer, is advantageous. Furthermore, new functional However, there still remains an urgent need for cost-effective
bioinks should be developed to allow high-speed printing and time-saving applications. Nevertheless, in the case of
using biocompatible materials with an acceptable rate of research, systems incorporating as many characteristics as pos-
degradation and immobilization of different types of growth sible should be devised. The principal idea which should be
factors. kept in mind while developing such models is “as complex
as necessary but as simple as possible”. 3D printing is a tech-
Outlook nology that stands out in this regard. It has the potential to
Utilization of 3D printing to construct a simple bone scaffold has mimic the healthy and diseased bone marrow, in varying
shown some successes in the area of bone graft fabrication, but degrees of complexities, tailored to the prerequisites of each
so far this was not the case for the models aimed to be used as a individual study. Having said that, this technique also faces
platform for expanding stem cells, or as in vitro models to test several challenges and further progress is necessary for
new treatment strategies or to study the different factors affecting successful practical applications.

12 ▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

Acknowledgments 15. V. Goncharova, N. Serobyan, S. Iizuka, I. Schraufstatter, A. de Ridder,


T. Povaliy, V. Wacker, N. Itano, K. Kimata, I.A. Orlovskaja,
We thank the publishers Elsevier, Taylor and Francis Ltd., Y. Yamaguchi, and S. Khaldoyanidi: Hyaluronan expressed by the hema-
BioMed Central, Springer Nature, John Wiley and Sons for the topoietic microenvironment is required for bone marrow hematopoiesis.
permission to reprint published data shown in Figs. 3(a)[47], 3 J. Biol. Chem. 287, 25419 (2012).
(b)[49], 3(c)[50], Figs. 4(a), 4(b)[124], 4(c)[121], 4(d), 4(e)[125]. 16. L. Coulombel, I. Auffray, M.H. Gaugler, and M. Rosemblatt: Expression
and function of integrins on hematopoietic progenitor cells. Acta
This project has received funding from the European Research Haematol. 97, 13 (1997).
Council (ERC) under the European Union’s Horizon 2020 17. C. Lee-Thedieck and J.P. Spatz: Biophysical regulation of hematopoietic
research and innovation programme (grant agreement No stem cells. Biomater. Sci. 2, 1548 (2014).
757490). C.L.-T. and A.R. acknowledge funding from the 18. M.R. Nelson and K. Roy: Bone-marrow mimicking biomaterial niches for
studying hematopoietic stem and progenitor cells. J. Mater. Chem. B 4,
BMBF NanoMatFutur programme (FKZ 13N12968) and the pro- 3490 (2016).
gramme BioInterfaces in Technology and Medicine (BIFTM) by 19. M.A. Walasek, R. van Os, and G. de Haan: Hematopoietic stem cell
the Helmholtz Association. expansion: challenges and opportunities. Ann. N. Y. Acad. Sci. 1266,
138 (2012).
20. H. Dombret and C. Gardin: An update of current treatments for adult
References acute myeloid leukemia. Blood 127, 53 (2016).
1. G.M. Crane, E. Jeffery, and S.J. Morrison: Adult haematopoietic stem cell 21. C. Nombela-Arrieta and S. Isringhausen: The role of the bone marrow
niches. Nat. Rev. Immunol. 17, 573 (2017). stromal compartment in the hematopoietic response to microbial infec-
2. H. Yoshihara, F. Arai, K. Hosokawa, T. Hagiwara, K. Takubo, tions. Front. Immunol. 7, 689 (2016).
Y. Nakamura, Y. Gomei, H. Iwasaki, S. Matsuoka, K. Miyamoto, 22. A. Knight: Animal experiments scrutinised: systematic reviews demon-
H. Miyazaki, T. Takahashi, and T. Suda: Thrombopoietin/MPL signaling strate poor human clinical and toxicological utility. Altex 24, 320 (2007).
regulates hematopoietic stem cell quiescence and interaction with the 23. E. Törnqvist, A. Annas, B. Granath, E. Jalkesten, I. Cotgreave, and
osteoblastic niche. Cell Stem Cell 1, 685 (2007). M. Öberg: Strategic focus on 3R principles reveals major reductions in
3. L.M. Calvi, G.B. Adams, K.W. Weibrecht, J.M. Weber, D.P. Olson, M. the use of animals in pharmaceutical toxicity testing. PLoS ONE 9,
C. Knight, R.P. Martin, E. Schipani, P. Divieti, F.R. Bringhurst, L. e101638 (2014).
A. Milner, H.M. Kronenberg, and D.T. Scadden: Osteoblastic cells regu- 24. J.S. Choi, B.P. Mahadik, and B.A.C. Harley: Engineering the hematopoi-
late the haematopoietic stem cell niche. Nature 425, 841 (2003). etic stem cell niche: frontiers in biomaterial science. Biotechnol. J. 10,
4. Y. Kunisaki, I. Bruns, C. Scheiermann, J. Ahmed, S. Pinho, D. Zhang, 1529 (2015).
T. Mizoguchi, Q. Wei, D. Lucas, K. Ito, J.C. Mar, A. Bergman, and P. 25. S. Mendez-Ferrer, T.V. Michurina, F. Ferraro, A.R. Mazloom, B.
S. Frenette: Arteriolar niches maintain haematopoietic stem cell quies- D. Macarthur, S.A. Lira, D.T. Scadden, A. Ma’ayan, G.N. Enikolopov,
cence. Nature 502, 637 (2013). and P.S. Frenette: Mesenchymal and haematopoietic stem cells form a
5. M. Acar, K.S. Kocherlakota, M.M. Murphy, J.G. Peyer, H. Oguro, C. unique bone marrow niche. Nature 466, 829 (2010).
N. Inra, C. Jaiyeola, Z. Zhao, K. Luby-Phelps, and S.J. Morrison: Deep 26. J. Kiernan, P. Damien, M. Monaghan, R. Shorr, L. McIntyre,
imaging of bone marrow shows non-dividing stem cells are mainly peri- D. Fergusson, A. Tinmouth, and D. Allan: Clinical studies of ex vivo
sinusoidal. Nature 526, 126 (2015). expansion to accelerate engraftment after umbilical cord blood trans-
6. I.G. Winkler, V. Barbier, B. Nowlan, R.N. Jacobsen, C.E. Forristal, J. plantation: a systematic review. Transfus. Med. Rev. 31, 173 (2017).
T. Patton, J.L. Magnani, and J.P. Levesque: Vascular niche E-selectin 27. N. Pineault and A. Abu-Khader: Advances in umbilical cord blood stem
regulates hematopoietic stem cell dormancy, self renewal and chemore- cell expansion and clinical translation. Exp. Hematol. 43, 498 (2015).
sistance. Nat. Med. 18, 1651 (2012). 28. J. Brandrup, E.H. Immergut, and E.A. Grulke: Polymer Handbook, 4th
7. M.J. Kiel, O.H. Yilmaz, T. Iwashita, O.H. Yilmaz, C. Terhorst, and S. ed. John Wiley and Sons: New York, 1999).
J. Morrison: SLAM family receptors distinguish hematopoietic stem 29. C.A. Muth, C. Steinl, G. Klein, and C. Lee-Thedieck: Regulation of hema-
and progenitor cells and reveal endothelial niches for stem cells. Cell topoietic stem cell behavior by the nanostructured presentation of extra-
121, 1109 (2005). cellular matrix components. PLoS ONE 8, e54778 (2013).
8. J. Fujisaki, J. Wu, A.L. Carlson, L. Silberstein, P. Putheti, R. Larocca, 30. A.J. Engler, S. Sen, H.L. Sweeney, and D.E. Discher: Matrix elasticity
W. Gao, T.I. Saito, C. Lo Celso, H. Tsuyuzaki, T. Sato, D. Cote, directs stem cell lineage specification. Cell 126, 677 (2006).
M. Sykes, T.B. Strom, D.T. Scadden, and C.P. Lin: In vivo imaging of 31. C. Lee-Thedieck and J.P. Spatz: Artificial niches: biomimetic materials
Treg cells providing immune privilege to the haematopoietic stem-cell for hematopoietic stem cell culture. Macromol. Rapid Commun. 33,
niche. Nature 474, 216 (2011). 1432 (2012).
9. A. Chow, D. Lucas, A. Hidalgo, S. Mendez-Ferrer, D. Hashimoto, 32. S.S. Kumar, J.H. Hsiao, Q.D. Ling, I. Dulinska-Molak, G. Chen, Y. Chang,
C. Scheiermann, M. Battista, M. Leboeuf, C. Prophete, N. van Rooijen, Y. Chang, Y.H. Chen, D.C. Chen, S.T. Hsu, and A. Higuchi: The combined
M. Tanaka, M. Merad, and P.S. Frenette: Bone marrow CD169 + macro- influence of substrate elasticity and surface-grafted molecules on the ex
phages promote the retention of hematopoietic stem and progenitor vivo expansion of hematopoietic stem and progenitor cells. Biomaterials
cells in the mesenchymal stem cell niche. J. Exp. Med. 208, 261 (2011). 34, 7632 (2013).
10. F. Arai, K. Hosokawa, H. Toyama, Y. Matsumoto, and T. Suda: Role of 33. C.C. Zhang and H.F. Lodish: Murine hematopoietic stem cells change their
N-cadherin in the regulation of hematopoietic stem cells in the bone surface phenotype during ex vivo expansion. Blood 105, 4314 (2005).
marrow niche. Ann. N. Y. Acad. Sci. 1266, 72 (2012). 34. Y. Zhou, H. Chen, H. Li, and Y. Wu: 3D culture increases pluripotent gene
11. M.A. Schroeder and J.F. DiPersio: Mobilization of hematopoietic stem expression in mesenchymal stem cells through relaxation of cytoskele-
and leukemia cells. J. Leukocyte Biol. 91, 47 (2012). ton tension. J. Cell. Mol. Med. 21, 1073 (2017).
12. G. Klein: The extracellular matrix of the hematopoietic microenviron- 35. L. Rodling, I. Schwedhelm, S. Kraus, K. Bieback, J. Hansmann, and
ment. Experientia 51, 914 (1995). C. Lee-Thedieck: 3D models of the hematopoietic stem cell niche
13. S.K. Nilsson, M.E. Debatis, M.S. Dooner, J.A. Madri, P.J. Quesenberry, under steady-state and active conditions. Sci. Rep. 7, 4625 (2017).
and P.S. Becker: Immunofluorescence characterization of key extracellu- 36. M.M. Cook, K. Futrega, M. Osiecki, M. Kabiri, A. Rice, K. Atkinson,
lar matrix proteins in murine bone marrow in situ. J. Histochem. G. Brooke, and M. Doran: Micromarrows—three-dimensional coculture
Cytochem. 46, 371 (1998). of hematopoietic stem cells and mesenchymal stromal cells. Tissue Eng.
14. K.D. Rodgers, J.D. San Antonio, and O. Jacenko: Heparan sulfate proteo- Part C 18, 319 (2012).
glycans: a GAGgle of skeletal-hematopoietic regulators. Dev. Dyn. 237, 37. E.C. Costa, D. de Melo-Diogo, A.F. Moreira, M.P. Carvalho, and I.
2622 (2008). J. Correia: Spheroids formation on non-adhesive surfaces by liquid

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪ 13
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
overlay technique: considerations and practical approaches. Biotechnol. 59. R. Jacamo, Y. Chen, Z. Wang, W. Ma, M. Zhang, E.L. Spaeth, Y. Wang,
J. 13, 1–12 (2018). V.L. Battula, P.Y. Mak, K. Schallmoser, P. Ruvolo, W.D. Schober, E.
38. F. Salamanna, D. Contartese, M. Maglio, and M. Fini: A systematic review J. Shpall, M.H. Nguyen, D. Strunk, C.E. Bueso-Ramos, S. Konoplev,
on in vitro 3D bone metastases models: a new horizon to recapitulate the R.E. Davis, M. Konopleva, and M. Andreeff: Reciprocal leukemia-stroma
native clinical scenario? Oncotarget 7, 44803–44820 (2016). VCAM-1/VLA-4-dependent activation of NF-kappaB mediates chemore-
39. A.M. Sitarski, H. Fairfield, C. Falank, and M.R. Reagan: 3d tissue engi- sistance. Blood 123, 2691 (2014).
neered in vitro models of cancer in bone. ACS. Biomater. Sci. Eng. 4, 60. Y. Saito, N. Uchida, S. Tanaka, N. Suzuki, M. Tomizawa-Murasawa,
324 (2018). A. Sone, Y. Najima, S. Takagi, Y. Aoki, A. Wake, S. Taniguchi, L.
40. J. Necas, L. Bartosikova, P. Brauner, and J. Kolar: Hyaluronic acid (hya- D. Shultz, and F. Ishikawa: Induction of cell cycle entry eliminates
luronan): a review. Vet. Med. 53, 397 (2008). human leukemia stem cells in a mouse model of AML. Nat.
41. B.D. Fairbanks, S.P. Singh, C.N. Bowman, and K.S. Anseth: Biotechnol. 28, 275 (2010).
Photodegradable, photoadaptable hydrogels via radical-mediated disul- 61. G.J. Cook, and T.S. Pardee: Animal models of leukemia: any closer to the
fide fragmentation reaction. Macromolecules 44, 2444 (2011). real thing? Cancer Metastasis Rev. 32, 63 (2013).
42. J. Zhang, A. Skardal, and G.D. Prestwich: Engineered extracellular matri- 62. M. de Jong, and T. Maina: Of mice and humans: are they the same?–
ces with cleavable crosslinkers for cell expansion and easy cell recovery. Implications in cancer translational research. J. Nucl. Med. 51, 501 (2010).
Biomaterials 29, 4521 (2008). 63. L. Demetrius: Of mice and men. EMBO Rep. 6, S39 (2005).
43. P.M. Kharkar, K.L. Kiick, and A.M. Kloxin: Designing degradable hydro- 64. F. Ishikawa, S. Yoshida, Y. Saito, A. Hijikata, H. Kitamura, S. Tanaka,
gels for orthogonal control of cell microenvironments. Chem. Soc. Rev. R. Nakamura, T. Tanaka, H. Tomiyama, N. Saito, M. Fukata,
42, 7335 (2013). T. Miyamoto, B. Lyons, K. Ohshima, N. Uchida, S. Taniguchi, O. Ohara,
44. F.R. Cheng, T. Su, J. Cao, X.L. Luo, L. Li, Y. Pu, and B. He: K. Akashi, M. Harada, and L.D. Shultz: Chemotherapy-resistant human
Environment-stimulated nanocarriers enabling multi-active sites for AML stem cells home to and engraft within the bone-marrow endosteal
high drug encapsulation as an “on demand” drug release system. region. Nat. Biotechnol. 25, 1315 (2007).
J. Mater. Chem. B 6, 2258 (2018). 65. C.R. Cogle, D.C. Goldman, G.J. Madlambayan, R.P. Leon, A.A. Masri, H.
45. C.M. Madl, B.L. LeSavage, R.E. Dewi, C.B. Dinh, R.S. Stowers, A. Clark, S.A. Asbaghi, J.W. Tyner, J. Dunlap, G. Fan, T. Kovacsovics,
M. Khariton, K.J. Lampe, D. Nguyen, O. Chaudhuri, A. Enejder, and S. Q. Liu, A. Meacham, K.L. Hamlin, R.A. Hromas, E.W. Scott, and W.
C. Heilshorn: Maintenance of neural progenitor cell stemness in 3D H. Fleming: Functional Integration of Acute Myeloid Leukemia into the
hydrogels requires matrix remodelling. Nat. Mater. 16, 1233 (2017). Vascular Niche. Leukemia 28, 1978 (2014).
46. T.M.A. Henderson, K. Ladewig, D.N. Haylock, K.M. McLean, and A. 66. L.J. Bray, M. Binner, Y. Korner, M. von Bonin, M. Bornhauser, and
J. O’Connor: Cryogels for biomedical applications. J. Mater. Chem. B C. Werner: A three-dimensional ex vivo tri-culture model mimics cell-cell
1, 2682 (2013). interactions between acute myeloid leukemia and the vascular niche.
47. A. Raic, L. Rodling, H. Kalbacher, and C. Lee-Thedieck: Biomimetic mac- Haematologica 102, 1215 (2017).
roporous PEG hydrogels as 3D scaffolds for the multiplication of human 67. A. Bruce, R. Evans, R. Mezan, L. Shi, B.S. Moses, K.H. Martin, L.
hematopoietic stem and progenitor cells. Biomaterials 35, 929 (2014). F. Gibson, and Y. Yang: Three-dimensional microfluidic tri-culture
48. M.S. Ferreira, W. Jahnen-Dechent, N. Labude, M. Bovi, T. Hieronymus, model of the bone marrow microenvironment for study of acute lympho-
M. Zenke, R.K. Schneider, and S. Neuss: Cord blood-hematopoietic stem blastic leukemia. PLoS ONE 10, e0140506 (2015).
cell expansion in 3D fibrin scaffolds with stromal support. Biomaterials 68. V. Trimarco, E. Ave, M. Facco, G. Chiodin, F. Frezzato, V. Martini,
33, 6987 (2012). C. Gattazzo, F. Lessi, C.A. Giorgi, A. Visentin, M. Castelli, F. Severin,
49. S.S. Kotha, B.J. Hayes, K.T. Phong, M.A. Redd, K. Bomsztyk, R. Zambello, F. Piazza, G. Semenzato, and L. Trentin: Cross-talk between
A. Ramakrishnan, B. Torok-Storb, and Y. Zheng: Engineering a multicel- chronic lymphocytic leukemia (CLL) tumor B cells and mesenchymal
lular vascular niche to model hematopoietic cell trafficking. Stem Cell. stromal cells (MSCs): implications for neoplastic cell survival.
Res. Ther. 9, 77 (2018). Oncotarget 6, 42130 (2015).
50. S. Sieh, A.A. Lubik, J.A. Clements, C.C. Nelson, and D.W. Hutmacher: 69. S. Tavor, I. Petit, S. Porozov, A. Avigdor, A. Dar, L. Leider-Trejo,
Interactions between human osteoblasts and prostate cancer cells in a N. Shemtov, V. Deutsch, E. Naparstek, A. Nagler, and T. Lapidot:
novel 3D in vitro model. Organogenesis 6, 181 (2010). CXCR4 regulates migration and development of human acute myeloge-
51. A.B. Bello, H. Park, and S.H. Lee: Current approaches in biomaterial- nous leukemia stem cells in transplanted NOD/SCID mice. Cancer Res.
based hematopoietic stem cell niches. Acta Biomater. 72, 1 (2018). 64, 2817 (2004).
52. Y.S. Torisawa, C.S. Spina, T. Mammoto, A. Mammoto, J.C. Weaver, 70. S.C. Mills, P.H. Goh, J. Kudatsih, S. Ncube, R. Gurung, W. Maxwell, and
T. Tat, J.J. Collins, and D.E. Ingber: Bone marrow-on-a-chip replicates A. Mueller: Cell migration towards CXCL12 in leukemic cells compared
hematopoietic niche physiology in vitro. Nat. Methods 11, 663 (2014). to breast cancer cells. Cell. Signal. 28, 316 (2016).
53. A. Reinisch, D.C. Hernandez, K. Schallmoser, and R. Majeti: Generation 71. B.-S. Cho, H.-J. Kim, and M. Konopleva: Targeting the CXCL12/CXCR4
and use of a humanized bone-marrow-ossicle niche for hematopoietic axis in acute myeloid leukemia: from bench to bedside. Korean J. Intern.
xenotransplantation into mice. Nat. Protoc. 12, 2169 (2017). Med. 32, 248 (2017).
54. C. Riether, C.M. Schurch, and A.F. Ochsenbein: Regulation of hemato- 72. A. Liou, C. Delgado-Martin, D.T. Teachey, and M.L. Hermiston: The
poietic and leukemic stem cells by the immune system. Cell Death CXCR4/CXCL12 axis mediates chemotaxis, survival, and chemoresist-
Differ. 22, 187 (2015). ance in t-cell acute lymphoblastic leukemia. Blood 124, 3629 (2014).
55. E. Stieglitz and M.L. Loh: Genetic predispositions to childhood leukemia. 73. E. Weisberg, A.K. Azab, P.W. Manley, A.L. Kung, A.L. Christie,
Ther. Adv. Hematol. 4, 270 (2013). R. Bronson, I.M. Ghobrial, and J.D. Griffin: Inhibition of CXCR4 in
56. T. Lapidot, C. Sirard, J. Vormoor, B. Murdoch, T. Hoang, CML cells disrupts their interaction with the bone marrow microenviron-
J. Caceres-Cortes, M. Minden, B. Paterson, M.A. Caligiuri, and J. ment and sensitizes them to nilotinib: potentiation of nilotinib by CXCR4
E. Dick: A cell initiating human acute myeloid leukaemia after transplan- antagonist. Leukemia 26, 985 (2012).
tation into SCID mice. Nature 367, 645 (1994). 74. E.L. Weisberg, M. Sattler, A.K. Azab, D. Eulberg, A. Kruschinski, P.
57. A.K. Brenner, I. Nepstad, and Ø. Bruserud: Mesenchymal stem cells sup- W. Manley, R. Stone, and J.D. Griffin: Inhibition of SDF-1-induced
port survival and proliferation of primary human acute myeloid leukemia migration of oncogene-driven myeloid leukemia by the L-RNA aptamer
cells through heterogeneous molecular mechanisms. Front. Immunol. 8, (Spiegelmer), NOX-A12, and potentiation of tyrosine kinase inhibition.
106 (2017). Oncotarget 8, 109973 (2017).
58. K. Schepers, E.M. Pietras, D. Reynaud, J. Flach, M. Binnewies, T. Garg, 75. Z.H. Shen, D.F. Zeng, X.Y. Wang, Y.Y. Ma, X. Zhang, and P.Y. Kong:
A.J. Wagers, E.C. Hsiao, and E. Passegue: Myeloproliferative neoplasia Targeting of the leukemia microenvironment by c(RGDfV) overcomes
remodels the endosteal bone marrow niche into a self-reinforcing leuke- the resistance to chemotherapy in acute myeloid leukemia in biomimetic
mic niche. Cell Stem Cell 13, 285 (2013). polystyrene scaffolds. Oncol. Lett. 12, 3278 (2016).

14 ▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
Prospective Article

76. T.M. Blanco, A. Mantalaris, A. Bismarck, and N. Panoskaltsis: The devel- Cells, Maria del Mar Vivanco, ed., Humana Press: New York, 2015;
opment of a three-dimensional scaffold for ex vivo biomimicry of human p. 213.
acute myeloid leukaemia. Biomaterials 31, 2243 (2010). 100.R. Dhurjati, V. Krishnan, L.A. Shuman, A.M. Mastro, and E.A. Vogler:
77. A.J. Favreau, C.P.H. Vary, P.C. Brooks, and P. Sathyanarayana: Cryptic Metastatic breast cancer cells colonize and degrade three-dimensional
collagen IV promotes cell migration and adhesion in myeloid leukemia. osteoblastic tissue in vitro. Clin. Exp. Metastasis 25, 741 (2008).
Cancer Med. 3, 265 (2014). 101.V. Krishnan, E.A. Vogler, D.M. Sosnoski, and A.M. Mastro: In vitro mim-
78. J.W. Shin and D.J. Mooney: Extracellular matrix stiffness causes system- ics of bone remodeling and the vicious cycle of cancer in bone. J. Cell.
atic variations in proliferation and chemosensitivity in myeloid leuke- Physiol. 229, 453 (2014).
mias. Proc. Natl. Acad. Sci. USA 113, 12126 (2016). 102.S. Bersini, J.S. Jeon, G. Dubini, C. Arrigoni, S. Chung, J.L. Charest,
79. T.T. Vu, C. Lim, and M. Lim: Characterization of leukemic cell behaviors M. Moretti, and R.D. Kamm: A microfluidic 3D in vitro model for specif-
in a soft marrow mimetic alginate hydrogel. J. Biomed. Mater. Res. B icity of breast cancer metastasis to bone. Biomaterials 35, 2454 (2014).
Appl. Biomater. 100, 1980 (2012). 103.T. Campbell, C. Williams, O. Ivanova, and B. Garrett: Could 3D printing
80. G.P. Gupta and J. Massagué: Cancer metastasis: building a framework. change the world. Technologies, Potential, and Implications of Additive
Cell 127, 679 (2006). Manufacturing, Atlantic Council, Washington, DC (2011).
81. I.J. Fidler: The pathogenesis of cancer metastasis: the “seed and soil” 104.S. Patra and V. Young: A review of 3D printing techniques and the future
hypothesis revisited. Nat. Rev. Cancer 3, 453 (2003). in biofabrication of bioprinted tissue. Cell Biochem. Biophys. 74, 93
82. F. Macedo, K. Ladeira, F. Pinho, N. Saraiva, N. Bonito, L. Pinto, and (2016).
F. Goncalves: Bone metastases: an overview. Oncol. Rev. 11, 321 (2017). 105.C. Arrigoni, M. Gilardi, S. Bersini, C. Candrian, and M. Moretti:
83. J. Massagué and A.C. Obenauf: Metastatic colonization by circulating Bioprinting and organ-on-chip applications towards personalized medi-
tumour cells. Nature 529, 298 (2016). cine for bone diseases. Stem Cell Rev. 13, 407 (2017).
84. C. Peitzsch, A. Tyutyunnykova, K. Pantel, and A. Dubrovska: Cancer stem 106.J.P. Temple, D.L. Hutton, B.P. Hung, P.Y. Huri, C.A. Cook,
cells: the root of tumor recurrence and metastases, Semin. Cancer Biol. R. Kondragunta, X. Jia, and W.L. Grayson: Engineering anatomically
44, 10 (2017). shaped vascularized bone grafts with hASCs and 3D-printed PCL scaf-
85. A.C. Obenauf and J. Massagué: Surviving at a distance: organ-specific folds. J. Biomed. Mater. Res. A 102, 4317 (2014).
metastasis. Trends. Cancer. 1, 76 (2015). 107.H.N. Chia and B.M. Wu: Recent advances in 3D printing of biomaterials.
86. A. Chambers, A. Groom, and I. MacDonald: Dissemination and growth of J. Biol. Eng. 9, 4 (2015).
cancer cells in metastatic sites. Nat. Rev. Cancer 2, 563a¯ (2002). 108.J. Groll, T. Boland, T. Blunk, J.A. Burdick, D.-W. Cho, P.D. Dalton,
87. G.D. Roodman and R. Silbermann: Mechanisms of osteolytic and oste- B. Derby, G. Forgacs, Q. Li, and V.A. Mironov: Biofabrication: reapprais-
oblastic skeletal lesions. Bonekey. Rep. 4, 1 (2015). ing the definition of an evolving field. Biofabrication. 8, 013001 (2016).
88. A.M. Sitarski, H. Fairfield, C. Falank, and M.R. Reagan: 3D tissue engi- 109.G. Gao, A.F. Schilling, T. Yonezawa, J. Wang, G. Dai, and X. Cui:
neered in vitro models of cancer in bone. ACS BACS Biomater Sci. Bioactive nanoparticles stimulate bone tissue formation in bioprinted
Eng. 4, 324 (2017). three‐dimensional scaffold and human mesenchymal stem cells.
89. D.T. Butcher, T. Alliston, and V.M. Weaver: A tense situation: forcing Biotechnol. J. 9, 1304 (2014).
tumour progression. Nat. Rev. Cancer 9, 108 (2009). 110.S. Catros, J.-C. Fricain, B. Guillotin, B. Pippenger, R. Bareille, M. Remy,
90. Y. Kang, P.M. Siegel, W. Shu, M. Drobnjak, S.M. Kakonen, E. Lebraud, B. Desbat, J. Amédée, and F. Guillemot: Laser-assisted bio-
C. Cordón-Cardo, T.A. Guise, and J. Massagué: A multigenic program printing for creating on-demand patterns of human osteoprogenitor cells
mediating breast cancer metastasis to bone. Cancer Cell 3, 537 (2003). and nano-hydroxyapatite. Biofabrication. 3, 025001 (2011).
91. Y. Shiozawa, E.A. Pedersen, A.M. Havens, Y. Jung, A. Mishra, J. Joseph, 111.C.-H. Chang, C.-Y. Lin, F.-H. Liu, M.H.-C. Chen, C.-P. Lin, H.-N. Ho, and
J.K. Kim, L.R. Patel, C. Ying, A.M. Ziegler, M.J. Pienta, J. Song, J. Wang, Y.-S. Liao: 3D printing bioceramic porous scaffolds with good mechan-
R.D. Loberg, P.H. Krebsbach, K.J. Pienta, and R.S. Taichman: Human ical property and cell affinity. PLoS ONE 10, e0143713 (2015).
prostate cancer metastases target the hematopoietic stem cell niche to 112.S. Wüst, M.E. Godla, R. Müller, and S. Hofmann: Tunable hydrogel com-
establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298 posite with two-step processing in combination with innovative hard-
(2011). ware upgrade for cell-based three-dimensional bioprinting. Acta
92. J. Insua-Rodríguez, and T. Oskarsson: The extracellular matrix in breast Biomater. 10, 630 (2014).
cancer. Adv. Drug Del. Rev. 97, 41 (2016). 113.K.-S. Hwang, J.-W. Choi, J.-H. Kim, H.Y. Chung, S. Jin, J.-H. Shim,
93. S. Nath and G.R. Devi: Three-dimensional culture systems in cancer W.-S. Yun, C.-M. Jeong, and J.-B. Huh: Comparative efficacies of
research: focus on tumor spheroid model. Pharmacol. Ther. 163, 94 collagen-based 3D printed PCL/PLGA/β-TCP composite block bone
(2016). grafts and biphasic calcium phosphate bone substitute for bone regen-
94. X. Xu, M.C. Farach-Carson, and X. Jia: Three-dimensional in vitro tumor eration. Materials (Basel) 10, 421 (2017).
models for cancer research and drug evaluation. Biotechnol. Adv. 32, 114.R. Alluri, A. Jakus, S. Bougioukli, W. Pannell, O. Sugiyama, A. Tang,
1256 (2014). R. Shah, and J.R. Lieberman: 3D printed hyperelastic “bone” scaffolds
95. T. Pan, E.L. Fong, M. Martinez, D.A. Harrington, S.-H. Lin, M. and regional gene therapy: a novel approach to bone healing. J.
C. Farach-Carson, and R.L. Satcher: Three-dimensional (3D) culture of Biomed. Mater. Res. A 106, 1104 (2018).
bone-derived human 786-O renal cell carcinoma retains relevant clinical 115.W. Zhang, Q. Lian, D. Li, K. Wang, D. Hao, W. Bian, J. He, and Z. Jin:
characteristics of bone metastases. Cancer Lett. 365, 89 (2015). Cartilage repair and subchondral bone migration using 3D printing
96. H. Kwon, H.J. Kim, W.L. Rice, B. Subramanian, S.H. Park, osteochondral composites: a one-year-period study in rabbit trochlea.
I. Georgakoudi, and D.L. Kaplan: Development of an in vitro model to BioMed Res. Int. 2014, 1 (2014).
study the impact of BMP‐2 on metastasis to bone. J. Tissue Eng. 116.W. Zhu, C. Xu, B.-P. Ma, Z.-B. Zheng, Y.-L. Li, Q. Ma, G.-L. Wu, and
Regen. Med. 4, 590 (2010). X.-S. Weng: Three-dimensional printed scaffolds with gelatin and plate-
97. R.F. Cox, A. Jenkinson, K. Pohl, F.J. O’Brien, and M.P. Morgan: lets enhance in vitro preosteoblast growth behavior and the sustained-
Osteomimicry of mammary adenocarcinoma cells in vitro; increased release effect of growth factors. Chin. Med. J. 129, 2576 (2016).
expression of bone matrix proteins and proliferation within a 3D collagen 117.S.V. Murphy and A. Atala: 3D bioprinting of tissues and organs.
environment. PLoS ONE 7, e41679 (2012). Nat. Biotechnol. 32, 773 (2014).
98. K.A. Fitzgerald, J. Guo, R.M. Raftery, I.M. Castaño, C.M. Curtin, 118.S. Adepu, N. Dhiman, A. Laha, C.S. Sharma, S. Ramakrishna, and
M. Gooding, R. Darcy, F.J. O’Brien, and C.M. O’Driscoll: Nanoparticle- M. Khandelwal: Three-dimensional bioprinting for bone tissue regener-
mediated siRNA delivery assessed in a 3D co-culture model simulating ation. Curr. Opin. Biomed. Eng. 2, 22 (2017).
prostate cancer bone metastasis. Int. J. Pharm. 511, 1058 (2016). 119.W. Peng, D. Unutmaz, and I.T. Ozbolat: Bioprinting towards physiolog-
99. R. Marlow and G. Dontu: Modeling the breast cancer bone metastatic ically relevant tissue models for pharmaceutics. Trends Biotechnol. 34,
niche in complex three-dimensional cocultures. In Mammary Stem 722 (2016).

MRS COMMUNICATIONS
Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms • www.mrs.org/mrc ▪ 15
of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203
120.P.S. Gungor-Ozkerim, I. Inci, Y.S. Zhang, A. Khademhosseini, and M.
R. Dokmeci: Bioinks for 3D bioprinting: an overview. Biomater. Sci 6,
915 (2018).
121.H. Cui, W. Zhu, M. Nowicki, X. Zhou, A. Khademhosseini, and L.
G. Zhang: Hierarchical fabrication of engineered vascularized bone
biphasic constructs via dual 3D bioprinting: integrating regional bioac-
tive factors into architectural design. Adv. Healthc. Mater. 5, 2174
(2016).
122.M. Hospodiuk, M. Dey, D. Sosnoski, and I.T. Ozbolat: The bioink: a com-
prehensive review on bioprintable materials. Biotechnol. Adv. 35, 217
(2017).
123.T. Jungst, W. Smolan, K. Schacht, T. Scheibel, and J. Groll: Strategies
and molecular design criteria for 3D printable hydrogels. Chem. Rev.
116, 1496 (2016).
124.H.W. Kang, S.J. Lee, I.K. Ko, C. Kengla, J.J. Yoo, and A. Atala: A 3D bio-
printing system to produce human-scale tissue constructs with struc-
tural integrity. Nat. Biotechnol. 34, 312 (2016).
125.W. Liu, Y.S. Zhang, M.A. Heinrich, F. De Ferrari, H.L. Jang, S.M. Bakht,
M.M. Alvarez, J. Yang, Y.-C. Li, G. Trujillo-de Santiago, A.K. Miri, K. Zhu,
P. Khoshakhlagh, G. Prakash, H. Cheng, X. Guan, Z. Zhong, J. Ju, G.
H. Zhu, X. Jin, S.R. Shin, M.R. Dokmeci, and A. Khademhosseini:
Rapid continuous multimaterial extrusion bioprinting. Adv. Mater. 29,
1604630 (2017).
126.X. Zhou, N.J. Castro, W. Zhu, H. Cui, M. Aliabouzar, K. Sarkar, and L.
G. Zhang: Improved human bone marrow mesenchymal stem cell oste-
ogenesis in 3D bioprinted tissue scaffolds with low intensity pulsed
ultrasound stimulation. Sci. Rep. 6, 1 (2016).
127.M.V. Braham, T. Ahlfeld, A.R. Akkineni, M.C. Minnema, W.J. Dhert, F.
C. Öner, C. Robin, A. Lode, M. Gelinsky, and J. Alblas: Endosteal and
perivascular subniches in a 3D bone marrow model for multiple
myeloma. Tissue Eng. Part C. Methods 24, 300 (2018).

16 ▪ MRS COMMUNICATIONS • www.mrs.org/mrc


Downloaded from https://www.cambridge.org/core. University of New England, on 02 Oct 2018 at 07:52:18, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
https://doi.org/10.1557/mrc.2018.203

You might also like