Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Sep 1.
Published in final edited form as: Adv Drug Deliv Rev. 2021 Jun 28;176:113852. doi: 10.1016/j.addr.2021.113852

Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro

Matthew L Tan 1,#, Lu Ling 1,#, Claudia Fischbach 1,2,#
PMCID: PMC8440401  NIHMSID: NIHMS1721829  PMID: 34197895

Abstract

Despite decades of research and advancements in diagnostic and treatment modalities, cancer remains a major global healthcare challenge. This is due in part to a lack of model systems that allow investigating the mechanisms underlying tumor development, progression, and therapy resistance under relevant conditions in vitro. Tumor cell interactions with their surroundings influence all stages of tumorigenesis and are shaped by both biological and biophysical cues including cell-cell and cell-extracellular matrix (ECM) interactions, tissue architecture and mechanics, and mass transport. Engineered tumor models provide promising platforms to elucidate the individual and combined contributions of these cues to tumor malignancy under controlled and physiologically relevant conditions. This review will summarize current knowledge of the biological and biophysical microenvironmental cues that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions and highlight advancements in tumor engineering approaches to further improve these technologies.

Keywords: Cancer heterogeneity, Extracellular matrix, Cancer metabolic transport, Tumor engineering, Tumor microenvironment, Tumor on a chip

1. Introduction

Cancer presents a major global healthcare challenge as it accounts for 1 in 6 deaths according to the World Health Organization. In the United States (US) alone, the National Cancer Institute (NCI) estimates that over 1,800,000 new cases and 600,000 deaths will result from the disease in 2021 [1]. Although great strides have been made in treating early-stage cancer, the survival rate of patients dramatically decreases in late-stage disease, which is characterized by metastasis and therapeutic resistance [1]. Given these connections, there is a clear clinical need for treatments that can prevent metastasis and poor clinical outcomes, but the development of such treatments requires improved understanding of the mechanisms underlying tumor development, progression, and therapy resistance.

Cancer progresses in a series of distinct phases: following initial growth, tumor cells migrate into the adjacent stroma, intravasate into blood vessels, circulate to distant sites where they extravasate and finally form secondary tumors [2]. This process can take decades and it is difficult to predict which cells disseminate from the main tumor and go on to form metastatic tumors. To further complicate matters, disseminated tumor cells may become dormant, enabling them to evade therapy and contribute to relapse [3]. However, the mechanisms behind tumor progression are poorly understood due to an almost exclusive focus on the tumor cells although it is now clear that the cancer’s evolving interactions with host tissues and the local microenvironment are equally essential [4]. Indeed, Stephen Paget already proposed in 1889 that cancer cells, which he referred to as “bad seeds”, need the right microenvironment, or “soil”, to metastasize effectively [5,6]. Since then, the tumor microenvironment has been shown abundantly to influence not only metastasis, but all stages of tumorigenesis [7,8]. In a pivotal study underscoring the importance of the microenvironment in regulating tumorigenesis, cancerous cells reversed their malignant phenotype when prevented from interacting with their surroundings [9]. In order to truly understand the mechanisms that drive cancer progression, research must not only focus on tumor cell-intrinsic features, but also consider the effects of their surrounding environment.

The tumor microenvironment is shaped by both biological and biophysical cues, which synergistically affect tumorigenesis (Figure 1) [10]. For example, cancer-associated fibroblasts, endothelial cells, and immune cells regulate the growth and invasion of tumors through paracrine signaling [10], while extracellular matrix (ECM) properties and changes in fluid flow impact tumorigenesis by varying the mechanical and transport properties of the microenvironment [11,12]. As cancer cells move through different tissue and organ systems the biochemical and biophysical cues to which they encounter vary considerably, and, simultaneously, the tumor cells themselves modify these cues to support their malignant progression [2]. Finally, genotypic and phenotypic variability between patients influences how tumor cells interact with their microenvironment, which contributes to interpatient tumor heterogeneity and complicates the development of effective therapies [13]. A better understanding of how tumor-microenvironment interactions and patient-specific changes thereof impact tumor initiation and progression will be necessary to develop more effective treatments and ultimately improve disease management.

Figure 1: Biological and biophysical hallmarks of the tumor microenvironment.

Figure 1:

Schematic of the cellular (cancer-associated fibroblasts [CAFs], endothelial cells, and immune cells) and acellular features (fibrotic remodeling of the extracellular matrix [ECM], metabolic/cytokine gradients, and interstitial fluid flow and pressure) of the tumor microenvironment. Increased recruitment of CAFs leads to compositional, structural, and mechanical changes of the ECM, while vascular dysfunction impacts mass transport of oxygen, nutrients, and paracrine signaling molecules. The latter results in varied gradient formation of solutes, elevated interstitial pressure and fluid flow as well as central necrosis. Collectively, these changes promote tumor progression and invasion where cancer cells evade immune surveillance, breach the basement membrane, migrate into the surrounding stroma, and eventually intravasate into an adjacent blood vessel to metastasize to distant organs. Image created with BioRender.com.

Although progress has been made in our mechanistic understanding of cancer, it is difficult to experimentally isolate the effect of specific microenvironmental conditions on human tumorigenesis with traditional models. Conventional monolayer culture does not recapitulate the 3D complexity of the microenvironment, while species-specific differences in tumorigenesis complicate translation of findings from mice to humans. Furthermore, mouse models do not allow for the use of human or patient-specific cells unless the animal is immune compromised, removing a major aspect of the tumor microenvironment. Engineered tumor models provide promising and constantly evolving alternatives that can be tailored to each experiment by adjusting their design parameters, including the utilized cell source, biomaterials, and fabrication technology. This review will summarize current knowledge of the biological and biophysical microenvironmental interactions that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions, and highlight advancements that could further improve these model systems. Ultimately, engineering-based cancer technologies provide innovative approaches to better understand how tumor-microenvironment interactions regulate cancer progression and enable screening of anti-cancer compounds under pathologically relevant conditions and in high-throughput settings.

2. Microenvironmental features regulating tumor progression

2.1. Cellular composition of the tumor microenvironment

2.1.1. Fibroblasts (and CAFs)

Cancer is often described as a wound that never heals and is associated with a sustained presence of reactive stromal fibroblasts termed cancer-associated fibroblasts (CAFs) [1416]. CAFs comprise a heterogeneous population of activated fibroblasts of different origins with elongated morphology and expression of markers such as α-smooth muscle actin (α-SMA) [17,18]. CAFs can promote tumorigenesis by causing both biochemical and biophysical changes to the microenvironment (Figure 1). Biochemical changes are mediated by a broad spectrum of CAF-secreted cytokines and chemokines. For example, CAF secretion of stromal cell-derived factor 1 (SDF1), hepatocyte growth factor (HGF), epidermal growth factor (EGF), and interleukin 6 (IL-6) induces tumor cell phenotypical changes including proliferation and stem-like properties through altered paracrine signaling [1922]. Biophysical changes in the microenvironment are introduced because CAFs are key drivers of fibrotic extracellular matrix (ECM) deposition and remodeling that represent hallmarks of most solid tumors (ECM) [18,23,24]. In particular, fibrotic ECM creates physical boundary conditions that directly and indirectly promote tumor progression by changing tumor cell behavior and by impeding drug delivery to desired sites, respectively, as described in more detail in section 2.2 below.

In addition to tumor cells, CAFs also influence other cell types. For example, CAFs can create immunosuppressive niches via secretion of IL-6 and transforming growth factor-β (TGFβ) [25] and promote the formation of dysfunctional vasculature by secreting vascular endothelial cell growth factor (VEGF) [26], which limits efficacious drug transport. Indeed, selective depletion of CAFs increases intratumoral drug uptake in breast and pancreatic cancers which, in turn, inhibits tumor growth and metastasis [2729]. Nevertheless, CAFs impact tumor progression in manifold and sometimes opposing ways depending on CAF populations and tumor stages. For example, depletion of alpha-smooth muscle actin (α-SMA) positive CAFs in early or late stage pancreatic cancer can worsen rather than improve tumor progression and survival [30]. These discrepancies underscore the need for relevant model systems that can help elucidate the divergent roles of CAFs in tumorigenesis.

2.1.2. Endothelial cells

In addition to transporting nutrients, metabolic byproducts, and drugs, blood vessels enable intra- and extravasation of tumor cells and provide perivascular niches that control tumor cell stemness and dormancy (Figure 1) [31,32]. Tumor interactions with the vasculature have been widely studied in the context of tumor angiogenesis, during which new blood vessels form from endothelial cells [33]. When exponential tumor growth leads to hypoxia, tumor and stromal cells upregulate angiogenic factors including VEGF, fibroblast growth factor-2 (FGF2), platelet derived growth factor (PDGF), angiopoietins, and matrix metalloproteinases (MMPs) to activate angiogenesis [3335]. In addition, dysregulated signaling pathways in tumor cells (e.g. overactive phosphoinositide 3-kinase [PI3K] signaling) alter endothelial cell behavior by increasing secretion of inflammatory cytokines such as IL-6 [36,37]. This combination of an overactivated angiogenic program and inflammatory response in endothelial cells results in blood vessels that are disorganized, leaky, and collapsed, leading to vascular dysfunction in the tumor microenvironment [38,39].

Aside from tumor-mediated effects on endothelial cells, endothelial cells reciprocally alter tumor cell behavior [4042]. For example, endothelial cell-secreted VEGF, angiopoietin 2 (ANGPT2), interleukin-8 (IL-8), IL-6, and TGFβ regulate the growth and cancer stem-like cell (CSC) characteristics of tumor cells [41,43,44]. This endothelial cell-mediated activation of stemness, in turn, enhances the ability of tumor cells to invade, initiate the formation of distant metastases, and evade drug treatment [4549]. Additionally, endothelial cells promote tumor cell dormancy and chemoresistance through direct cell contact [42,50,51]. Given these connections, engineered tumor models should be designed in a manner that allows the investigation of pheno- and genotypic changes in both tumor and endothelial cells simultaneously.

2.1.3. Immune cells

Immune cells are another key component of the microenvironment (Figure 1), and tumor associated macrophages (TAMs) are one of the most extensively studied of these cell types [52]. Depending on the particular subpopulation (e.g., M1-biased or M2-biased macrophages as two extreme examples, although there is a much broader range of phenotypes), TAMs can have either pro- or anti-inflammatory effects. While anti-inflammatory macrophages are most widely studied for their tumor-promoting effects, pro-inflammatory macrophages can similarly stimulate malignancy [53]. For example, anti-inflammatory TAMs express cytokines and chemokines (e.g. C-C motif chemokine ligand 2 [CCL2], C-X-C motif chemokine ligand 12 [CXCL12], IL-6, interleukin-1 beta [IL-1β], colony stimulating factor-1 [CSF-1], and VEGF) that can suppress antitumor immunity while promoting tumor progression [54]. This has been manifested in TAMs increasing tumor cell migration, invasion, and metastasis [5557]. TAMs can further drive tumor growth and metastasis by altering other stromal cell types that, for example, induce tumor angiogenesis [58,59]. In addition to TAMs, other immune cells such as T cells and natural killer (NK) cells are significant contributors to the immune characteristics of the tumor microenvironment and are discussed in more detail elsewhere [60]. More generally, the tumor microenvironment often exhibits characteristics of an immunosuppressive milieu, as cancer cells and associated stromal cells express negative immune regulators such as checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) [61]. In addition to these biochemical mediators, escape from immune surveillance can occur due to biophysical mechanisms. For example, dense layers of fibrotic ECM and dysfunctional vasculature often inhibit infiltration of immune effector cells into more central tumor regions [62,63], which may be further aggravated by the fact that immune cells can activate fibroblasts and thus, ECM remodeling [64]. Better understanding the diverse roles of immune cells as a function of biochemical and biophysical heterogeneity within the tumor microenvironment has the potential to broaden the effectiveness of immunotherapies.

2.2. Acellular and biophysical aspects of the tumor microenvironment

2.2.1. ECM – Composition, structure, mechanics

In healthy tissues, epithelial cells interact with the basement membrane (BM), a sheet like, nanoporous layer of ECM primarily composed of collagen type IV and laminins [65]. The BM contributes to the barrier function of epithelial tissues and is critical for their development, organization, and polarization [65,66]. However, during malignant transformation, cells break down the BM either proteolytically or mechanically to enable tumor invasion [65,66]. Tumor-associated fibrotic remodeling by CAFs (also termed desmoplasia) is characterized by increased deposition of collagen type I, III, and VI, fibronectin, and hyaluronan (HA) [18,64,67]. These changes can be detected clinically through mammographic screening and regulate the behavior of both tumor and stromal cells by varying adhesion-dependent cell signaling. For example, increased collagen I levels drive tumor cell invasion and metastasis by activating integrins and collagen I receptor discoidin domain receptor 2 (DDR2) [68,69], while increased fibronectin promotes tumor cell invasion and stromal cell secretion of proangiogenic factors in an integrin-dependent manner [70,71]. Changes in HA content regulate tumor cell stemness and interactions with the vasculature through CD44, but can also impact integrin signaling with consequences on invasion and metastasis [7274]. Finally, changes in ECM composition alter biochemical signaling within the tumor microenvironment by influencing the sequestration, binding, and release of many bioactive molecules [23,75] including VEGF and FGF-2 to promote angiogenesis and tumor cell proliferation [76,77].

Compositional changes simultaneously alter the microarchitecture and mechanical properties of the ECM with functional consequences on tumor progression (Figure 1). In particular, stromal ECM consists of a porous network of fibers whose structure changes in the presence of a tumor [78]. While collagen fibers in healthy breast tissue are thin, relaxed, and curvy, fibers in tumors are thick, stretched, and straight [79,80]. Furthermore, fibers align parallel to the tumor and help prevent invasion during benign stages of cancer, while more advanced stages are characterized by fibers aligning perpendicular to the tumor providing guidance cues for invasion [79,81]. Importantly, collagen fiber alignment increases ECM stiffness, which promotes tumor malignancy through positive mechanical feedback as greater ECM stiffness increases cell traction forces mediating more fiber alignment and tumor progression [82]. While some of these changes are reversible, cross-linking through matrix processing enzymes such as lysyl oxidase (LOX) increases ECM stiffness irreversibly and influences cell behaviors via changes in mechanosignaling [69,83].

Cells translate altered ECM stiffness through mechanoreceptors including integrins, which alter gene expression by stimulating Rho/ROCK-mediated cell contractility, and activating the transcription factors YAP/TAZ [8486]. These changes result in loss of cell polarity, reduced cell-cell adhesion, and increased proliferation ultimately contributing to metastatic invasion [85,87,88]. Importantly, stiffness-induced changes in cell contractility can influence the bioactivity of several ECM components as well as soluble signaling molecules. For example, cellular traction partially unfolds fibronectin with consequences on its bioactivity and fiber stiffness as well as subsequent deposition of collagen [89,90] and mechanoactivation of TGF-β signaling [91]. In summary, changes in tumor-associated ECM composition, structure, and mechanics collectively regulate cancer progression and thus, should guide the design of engineered tumor models.

2.2.2. Mass transport – Metabolism, hypoxia, and perfusion

In the tumor microenvironment, dysfunctional vasculature, lymphatics, and aberrant ECM remodeling, result in the disruption of mass transport homeostasis [92]. In contrast to normal vasculature, blood vessels in tumors are disorganized, leaky, and collapsed, leading to a unique spatiotemporal distribution of nutrients, oxygen, and metabolites (Figure 1) [38,93]. In combination with the forces exerted by the expansion of solid tumors and lack of fluid clearance due to impaired lymphatic function, mass transport due to convection and diffusion is further perturbed by abnormal interstitial pressure and fluid flow in the tumor microenvironment [94]. The metabolic and physical effects caused by this unique transport situation have been demonstrated to increase the malignancy of tumor cells [92].

One of the most extensively studied consequences of perturbed mass transport is hypoxia, which regulates all stages of cancer by stabilizing the transcriptional regulator hypoxia-inducible factor subunit 1α (HIF-1α) [9597]. For example, hypoxia/HIF-1α upregulates dormancy markers in primary tumors, suggesting that transport limitations can pre-determine the fate of tumor cells in metastatic target organs even prior to invasion and dissemination [98]. By promoting ECM remodeling and the expression of integrins, hypoxia also contributes to the compositional and mechanical changes of tumor-associated ECM described above, which then work in concert to increase the metastatic potential of tumor cells [99101]. Because of the importance of hypoxia in modulating cancer progression, therapeutic strategies to target HIF-1α or normalize blood vasculature have been proposed, and discoveries related to HIF-1α have been awarded a Nobel prize [37,102,103]. While HIF-1α is the most widely studied mediator of hypoxia responses, it should be noted that other signaling mechanisms including HIF-2α and the nuclear factor-κB (NF-κB) pathway can also mediate responses to hypoxia in tumor cells [104,105].

Mass transport-dependent spatial variations in hypoxia are also essential regulators of tumor metabolic reprogramming and thus, heterogeneity [93,106]. For example, the core of tumors is characterized by a decrease in glucose and oxygen and an increase in waste products such as lactate [107]. While a lactate-dependent reduction of pH can globally promote tumor progression [108], spatial variations thereof alter the metabolic cross-talk between cells in different regions of the tumor. More specifically, hypoxic cancer cells in the core generate excess lactate from upregulated glycolysis. This lactate can then utilized as a metabolic substrate to sustain the anabolic needs of oxygenated cancer cells at the periphery of the tumor, ensuring tumor growth and survival [93,109]. Similar scenarios also occur between cancer and stromal cells, as CAFs can support the growth and progression of tumor cells in a reverse Warburg effect during which they produce lactate by aerobic glycolysis that is then used by tumor cells [110]. It is worth noting that in addition to fulfilling energy needs, the presence of lactate, as well as other metabolites such as glutamine and asparagine, independently regulate tumor malignancy and aggressiveness [111,112].

Another feature of altered mass transport in the tumor microenvironment manifests in elevated interstitial fluid pressure, often resulting in fluid flow towards lymphatic vessels [94,113]. This imparts shear forces and mechanical stress on tumor cells which modulate their invasive and malignant potential [114]. For example, elevated interstitial pressure can promote collective invasion through increased tumor cell expression of markers involved in the epithelial-mesenchymal transition (EMT) such as Snail, vimentin, and E-cadherin [115]. Similarly, interstitial flow enhances tumor cell invasion and intravasation through factors such as flow direction (transmural vs. luminal with regard to lymphatic vessels), flow strength, as well as through the redistribution of chemokine gradients [116118]. The effects of fluid flow are not limited to the tumor cells, but also impact the behavior of stromal cells including fibroblasts and macrophages, which can then independently enhance tumor cell invasion [119,120]. These results underscore the significance of including the physical traits of mass transport when studying tumor-microenvironment interactions.

3. Engineered tumor microenvironments

3.1. Model systems recapitulating the cellular complexity of tumors

3.1.1. Conventional in vitro approaches to study tumorigenesis

Preclinical models have been used for decades to model tumor progression, and animal studies remain the gold standard for screening potential drug candidates. However, many challenges remain as most animal models are expensive and the spatiotemporal dynamics of tumor progression are not easily identified or visualized, complicating mechanistic understanding. In vitro systems enable improved control over experimental variables while maintaining aspects of physiological relevance. Historically, the most commonly used in vitro models to study cancer are immortalized cell lines growing as adherent 2D cultures. These cultures are readily accessible, easy to maintain, and can be scaled up for rapid drug screenings. However, many drug candidates fail in clinical trials, despite promising in vitro and in vivo data [121]. This lack of predictive power and other gaps in performance during translational research are linked to many factors [122]. One important contributor is the lack of cellular heterogeneity that is a hallmark of tumors in vivo, in typical in vitro models [123,124]. For example, cell lines grown in conventional 2D culture are intrinsically homogenous, which is further reinforced by the lack of cellular and acellular microenvironmental cues described in the previous sections. In contrast, 3D model systems can integrate these components and thus provide a suitable intermediate between 2D cell culture and in vivo models that can mimic some of the intratumor heterogeneity impacting clinical prognosis. When developing physiologically relevant 3D models of the tumor microenvironment, several design parameters need to be considered: selection of cellular components, choice of relevant ECM materials, generation of fluid flow, and biochemical gradients, etc. These design principles and examples for corresponding models will be discussed in the following sections.

3.1.2. Multicellular spheroid cultures

Spheroid cultures refer to multicellular aggregates mimicking 3D cell-cell adhesions found in vivo [125,126]. Spheroids are most easily formed from cell lines using the “hanging drop” method, which involves adding a droplet of cell suspension on the lid of a tissue culture plate, which is then inverted to encourage cell-cell adhesion and spheroid formation due to surface tension [127]. However, this approach often results in aspherical aggregates, which need to be transferred to a secondary culture vessel. Both limitations can be circumvented by direct culture on non-adherent surfaces such as agar, agarose or ultra-low attachment culture plates with predefined well geometry (spherical or tapered) [128] and further improved by the addition of rotation culture and viscous media (cellulose) [129]. More recently, advanced micro-patterning techniques and microfluidic devices have been demonstrated to yield the large quantities of consistently sized spheroids necessary for large-scale drug screening [130132]. The drawback to these methods is increased cost and labor-intensive maintenance.

Spheroid cultures provide 3D cell-cell communication and physiologically relevant structures that resemble in vivo tumor conditions [133]. As cells grow and spheroids expand in size, diffusion of nutrients and oxygen throughout the spheroid becomes limited, leading to nutrient starvation and central hypoxia. Indeed, spheroids > 400–500 μm in diameter exhibit characteristic zones of peripheral proliferating cells and necrotic centers mimicking the pathophysiological gradients found in vivo (Figure 2A)[128,134]. In addition to mono-culture spheroids, heterotypic cell–cell interactions can be recapitulated by co-culturing cancer and stromal cells [135]. Co-encapsulating cancer cells, CAFs, and monocytes in a single spheroid, for example, recapitulates tumor-immune compartments by driving monocytes to adopt a TAM-like phenotype [136]. Furthermore, this heterotypic spheroid model exhibited increased resistance to paclitaxel compared to mono-cultures, suggesting that reciprocal interactions between these different cellular components alter drug sensitivity and thus, should be incorporated into models used for in vitro drug testing (Figure 2B)[136].

Figure 2: Multicellular spheroid and organoid models.

Figure 2:

A. Multicellular spheroids formed from a single cancer cell line recapitulate diffusion limited transport resulting in spatially distinct regions of proliferation and necrosis that mimic the pathophysiological heterogeneity of tumors in vivo. Reproduced from Impact Journals, LLC with an open-access copyright policy. [134]. B. Co-culture spheroids containing tumor cells (red), CAFs, and monocytes recapitulate stromal heterogeneity. Cisplatin treatment caused rapid apoptosis (green) in these cultures, while paclitaxel was less effective. Reproduced from Elsevier Ltd with an open-access copyright policy. [136]. C. Mouse pancreatic stellate cells (PSCs, red) and pancreatic cancer organoids (green) can be embedded in Matrigel to increase stromal populations. PSCs cultured in Matrigel alone remain quiescent, but PSCs in contact with tumor organoids assume CAF-like phenotypes. Reproduced with permission from Rockefeller University Press [139]. D. Different types of patient-derived tumor organoids (bottom row) can be formed via the air–liquid interface (ALI) method, a technique that recapitulates parental tumor histology (top row) and maintains immune cell populations. Reproduced with permission from Elsevier Ltd [143].

3.1.3. Organoid cultures

Patient-derived organoid cultures provide experimental platforms to recapitulate the cellular heterogeneity of tumors in patients. Tumor organoids are derived from dissociated tissue samples or circulating tumor cells that are encapsulated in bio-mimetic matrix to encourage self-assembly and differentiation into more complex structures [137]. The most commonly used method to generate organoids is to embed samples into Matrigel®, a BM extract derived from mouse Engelbreth-Holm-Swarm (EHS) sarcoma which preserves the heterogeneity of cancer cell populations [138]. Other stromal components such as CAFs [139], immune cells [140], and vascular networks [140] can be cultured together with tumor organoids to increase cellular complexity and further assess cell-cell interactions within the tumor microenvironment (Figure 2C) [139]. Air–liquid interface (ALI) culture is another method of forming organoids in which tissues fragments are embedded into collagen with one side facing the media and the other facing air via a transwell membrane [141,142]. The enhanced oxygenation afforded by this method improves the conservation of stromal populations from the original tissues (Figure 2D) [143]. To aid in the use of these models, tumor organoid bio-banks have been generated from patient-derived and mouse-derived tumor specimens as well as matching normal tissues, enabling more widespread applications of patient samples in research [138,144].

Organoid cultures enable biomarkers and other morphological changes to be monitored and measured across different time scales ex vivo, providing unprecedented insight into the dynamics of biological processes [137,145,146]. This is particularly useful for tumorigenesis studies and patient-specific drug screening, where matching organoids can be generated to compare cellular responses in tumor and healthy tissue [147]. In addition, pluripotent stem cell-derived normal tissue organoids can be modified via CRISPR-Cas9 genome editing to become tumor organoids, which provide a useful tool to track and decipher the genetic determinants of tissue morphogenesis and cancer progression [148]. However, the use of organoid cultures may be complicated due to variability resulting from tissue harvesting and formation efficiency. Moreover, expansion of organoids may lead to selective loss or overgrowth of stromal populations, limiting studies focused on tumor-specific heterogeneity [143,149,150]. Another limitation is the use of complex media that requires specific supplements and frequently uses fetal bovine serum, which could potentially influence cell signaling and drug efficacy testing, but could be overcome by using matched patient peripheral blood serum [151,152]. Not all organoid cultures can be expanded long term and tumor organoid biobanks are primarily focused on epithelial cancers, although tumor organoids can be generated from other cancers such as glioblastoma [153]. Collectively, organoid cultures are an invaluable technology to create patient-specific 3D in vitro models of cancer and are likely to replace cell line studies in the future.

3.2. Biomaterials-based model systems recapitulating tumor-ECM interactions

3.2.1. Natural biomaterials

Embedding cells into biomaterials mimicking the compositional and biophysical properties of tumor-associated ECM allows studying the resulting functional consequences on tumorigenesis. Given their intrinsic bioadhesivity, natural biomaterials isolated from mammalian tissues are particularly attractive. BM-mimicking materials such as Matrigel® and Cultrex® are widely used to study organotypic tissue morphogenesis and initial invasion of transformed epithelial cells through the BM [154156]. For example, normal epithelial cells cultured in Matrigel® adopt spherical and acinar structures similar to mammary tissues, but activation of oncoproteins such as papillomavirus E7 (HPV E7) and erythroblastic oncogene B (ErbB2) can result in excessive proliferation and aberrant morphogenesis (Figure 3A)[156]. While Matrigel® mimics the biochemical composition of the BM, it is subject to batch-to-batch variations and lacks the structural and mechanical properties of the BM in vivo [157]. This shortcoming can be partially circumvented by using BM in its native and intact form (e.g., isolated from rat peritoneum), but limited quantities usually prevent large scale experiments [157,158]. In addition, commercial BM preparations cannot recapitulate the range of tissue stiffnesses present in normal versus diseased tissues (e.g. 10 times stiffer in breast cancer vs. normal breast tissue [85]) or different metastasis-prone organs (hundreds of pascals in brain versus gigapascals in bones [159,160]). Furthermore, commercial BM preparations contain growth factors and cytokines that affect cell behavior independently. One approach to avoid these limitations is to use single ECM components including collagen [82,161], fibronectin [162], and HA [163,164].

Figure 3: Biomaterials models to study cell-ECM interactions.

Figure 3:

A. Culture in Matrigel® can be used to test the effect of malignant transformation on morphogenesis. Normal epithelial cells (MCF10A) form small acinar structures similar to human mammary tissues (top right) when cultured in Matrigel®, while transformed cells (human papillomavirus E7 [HPV E7] and erythroblastic oncogene B [ErbB2]) are larger and compose multiacinar structures. Reproduced with permission from Elsevier Ltd. [156]. B. (i) When embedded into collagen, co-culture spheroids composed of tumor cells and obesity-associated stromal cells invade collectively due to collagen displacement, a process that can be partly inhibited by MMP inhibition with Batimastat. Reproduced with permission from John Wiley & Sons, Inc. [135]. (ii) Culturing breast cancer cells in aligned collagen fiber matrices induced morphological changes (top images, cells in green, collagen in grey) that led to increased directional migration (bottom image, cells in cyan). Reproduced with permission from Elsevier [178]. C. Decellularized ECM can be derived from fibroblasts cultured in vitro. ECM derived from alpha-SMA (red) positive, desmoplastic human fibroblasts is more aligned and enriched for fibronectin (green) compared to ECM derived from normal fibroblasts. Reproduced with permission from Elsevier [182]. D. A hybrid alginate hydrogel modified with PEG spacers enables introduction of varied viscoelastic properties. Faster stress relaxation (left to right) promoted osteogenic differentiation of mesenchymal stem cells as indicated by increased matrix mineralization (Von Kossa staining, top panel) and type-1 collagen deposition (bottom panel). Reproduced with permission from Springer Nature [203]. E. Electrospun networks of RGD-modified dextran methacrylate (DexMA) fibers permit independent tuning of fiber and bulk mechanical properties and revealed that soft fiber stiffness (top panel) promotes cell spreading (cells outlined in magenta) typically associated with stiff matrices by enabling cellular recruitment of individual fibers. Reproduced with permission from Springer Nature [215].

Type I collagen has been extensively used to form defined ECM hydrogels. Collagen type I-based hydrogels have increased our understanding of how ECM in the tumor microenvironment regulates stromal cells [82,165], tumor cell proliferation and migration [166,167], and the collective behavior of both of these cell types (Figure 3B.i) [135]. These insights would not have been possible with BM-mimicking materials as cells embedded into Matrigel® undergo morphogenetic growth, while they exhibit protrusive migration in collagen gels [168]. The ability to adjust the mechanical and structural properties of collagen hydrogels via changes in concentration [169], adjustment of gelation parameters (e.g. temperature and pH) [82,169], as well as glycation [170,171] presents another advantage. Importantly, varying the gelation parameters simultaneously alters the fibrillar structure of collagen [172], which impacts the viscoelastic behavior of the resulting gels and enables cell-mediated local strain-stiffening through positive mechanical feedback [82]. The fibrillar structure of collagen can also be controlled via electrospinning [173], by applying a magnetic field [174], using guided cellular compaction [175], or through mechanical stretching [176] and shear flow [177]. For example, to study the effect of fiber alignment on tumor cells at the invasion front, one study used a strain device to create linearized ECM and showed that fiber alignment, rather than stiffness, promotes directional migration of breast cancer cells (Figure 3B.ii) [178].

While single ECM components such as collagen provide invaluable tools, natural ECM usually comprises many different ECM proteins that are biochemically and biophysically distinct. The compositional, mechanical, and architectural complexity introduced by these multiple ECM components synergistically regulate cellular behavior and are challenging to recapitulate in more defined ECM cultures. To evaluate how the ECM of specific stromal cell types affects tumorigenesis under conditions mimicking the compositional and mechanical complexity of native ECM, decellularized cell-derived ECMs are increasingly used. For example, CAF-derived ECMs retain desmoplastic features in vitro and drive cancer proliferation and drug resistance relative to ECMs derived from normal fibroblasts (Figure 3C) [179182]. Similar approaches have also revealed that ECM changes drive tumorigenesis by altering the behavior of tumor and immune cells in the context of obesity, a condition known to be epidemiologically associated with increased risk and worse prognosis of cancer patients [183,184]. In the context of bone metastasis, decellularized ECMs produced by osteoblasts can mimic aspects of mineralized collagen, which is the basic building block of bone ECM and regulates the behavior of several bone-tropic cancer cell types including prostate and breast cancer cells [185,186]. To retain the compositional and architectural properties of native ECM even more fully, whole tissues or organs can be decellularized [67,187]. Indeed, this approach has been used for regenerative medicine applications [188], proteomic mapping of ECM composition [67,189], encapsulation of organoid cultures [190,191], and also offers promise for tumor engineering applications. For example, colorectal cancer cells seeded in decellularized ECM from liver and lungs can be used to model organ specific metastasis [190]. However, it is important to note that decellularization protocols influence which ECM components are retained, complicating comparisons across different studies [188]. Furthermore, the complexity of these models makes it difficult to isolate how specific compositional or physical properties of the ECM influence cancer behaviors.

3.2.2. Synthetic and composite biomaterials

Synthetic materials offer promising alternatives to natural biomaterials given their highly defined nature, reproducibility, and readily adjustable structural and mechanical properties. In particular, polyacrylamide (PA) hydrogels have been widely used to study tumorigenesis as a function of ECM stiffness as they can be designed to exhibit a wide range of stiffnesses while maintaining constant surface chemistry [192,193]. However, PA gels cannot be used for 3D culture studies due to cytotoxicity of their monomers and the polymerization procedure itself [188]. Polyethylene glycol (PEG) gels help circumvent this limitation, but these materials need to be covalently modified with adhesion ligands such as RGD found in fibronectin [194] or GFOGER in collagen [195] and/or MMP-degradable moieties [196] to enable cellular interactions reminiscent of native ECM. Alternatively, PEG hydrogels can be cross-linked with natural proteins such as HA to readily incorporate adhesion and degradation sites [197,198]. These modifications enable studies investigating the roles of ECM stiffness, cell adhesion, and migration [197200].

Despite the highly versatile functions of synthetic ECM models, one major limitation is that synthetic hydrogels composed of only PA and PEG are typically linearly elastic and thus, do not capture the stress-relaxation properties of native viscoelastic interstitial ECM [201]. Nevertheless, it should be mentioned that it is possible to create viscoelastic PA gels by entrapping a linear PA solution in an elastic network of PA hydrogels [17]. Another complementary approach is to use PEG hydrogels cross-linked by reversible hydrazone bonds [202]. The dynamic stress-relaxing crosslinks in these systems permit complex cellular functions to occur while retaining the benefits of traditional covalently crosslinked hydrogels. Alternatively, it is possible to combine synthetic approaches with natural biomaterials in composite systems. For example, one system utilized ionically-crosslinked alginate hydrogels coupled with PEG spacers to enable introduction of varied viscoelastic properties independent of the initial elastic modulus and degradation [203]. It was then demonstrated that faster stress relaxation promoted fibroblast cell spreading and proliferation, and osteogenic differentiation of mesenchymal stem cells in this 3D hydrogel (Figure 3D) [203]. Being able to dynamically tune the viscoelastic properties of hydrogels provides additional avenues to study how ECM mechanics affect tumorigenesis.

In addition to hydrogels, synthetic materials such as poly(lactide-co-glycolide) (PLG), polylactic acid (PLA), poly(ester urethane) (PUR), and poly(ε-caprolactone) (PCL) can be fabricated using different strategies including phase separation, gas-foaming/particulate leaching and electrospinning to create porous scaffolds that are biocompatible and amenable to large-scale use [204208]. Indeed, cancer cells seeded into porous PLG scaffolds formed tissues that mimic tumor histological features [209]. Furthermore, limited oxygen and nutrient transport in these engineered tumors led to increased malignancy compared to the same cells cultured in conventional 2D culture or 3D Matrigel [209]. The biophysical properties of PLG, PCL, and PUR scaffolds can be advantageous as they can achieve stiffnesses in the MPa range while providing substrates for mineralization that mimic the inorganic composition of skeletal ECM at bone metastatic sites [210212]. In addition, implantation of microporous PCL scaffolds into mice promotes infiltration of circulating tumor and immune cells, and therefore creates a metastatic niche that can be sampled via biopsy to study disease progression and treatment response [213]. On the other hand, electrospun PCL fibrillar scaffolds can recapitulate aligned ECM substrates and have demonstrated that ECM alignment promotes epithelial-mesenchymal transition of breast cancer cells that is relevant to tumor invasion [214]. Interestingly, another study using electrospun dextran methacrylate (DexMA) fibers to independently tune individual fiber and bulk matrix mechanical properties indicated that softer fibers induce cell spreading and proliferation due to increased fiber recruitment and focal adhesion formation (Figure 3E) [215]. This finding was in contrast to previous observations using non-fibrous hydrogels and highlighted the importance of biomaterial structure in regulating cell behaviors. Despite their many advantages, synthetic scaffolds cannot be remodeled by cells and are subject to polymer degradation, which in the case of PGA and PLA result in the release of acidic byproducts that can independently influence cell behavior [216]. Collectively, biomaterials scaffolds can be tailored to recapitulate the compositional, structural, and mechanical properties of tumor-relevant ECM to study its impact on tumorigenesis, but careful consideration should be given to their choice of materials and fabrication method.

3.3. Microfabricated models to study transport considerations

3.3.1. Models to generate gradients and interstitial pressure

To understand how soluble factor gradients influence tumor progression, model systems are necessary to precisely control soluble factor delivery, interstitial flow, and pressure (Figure 4A). Microfluidic platforms made of polydimethylsiloxane (PDMS) are most commonly utilized to model physiological transport mechanics and generate controlled chemical gradients in a source and sink configuration by allowing modulation of geometries, materials, and inputs [217,218]. Such gradient generating microfluidics have enabled studies of cancer cell invasion and chemotaxis in response to exogenous or cell-generated chemokine gradients in both 2D and 3D contexts [219223]. More recently, microfluidic models have been used to study the effects of metabolic limitations in tumors, including hypoxia and nutrient/waste gradients where nutrients are introduced in a single channel and gradients are generated intrinsically by cells seeded adjacent to the channel [224,225]. Such models demonstrated that tumor cells alter their metabolism and gene expression signatures in response to nutrient and oxygen gradients [224,225]. Recent work has utilized alternative methods to microfluidics, such as an O2-controllable hydrogel that is able to generate hypoxic gradients without the need for complex microfabrication and flow setups [226,227]. Other innovative approaches to study the effect of chemokine or metabolic gradients on cancer include work from the McGuigan lab, which has pioneered a platform known as TRACER. In this work, cells are seeded onto a bio-composite strip that is then rolled onto an oxygen impermeable core, generating a naturally formed hypoxic gradient (Figure 4B) [228]. After culture, the strip can be unrolled to enable selective characterization of cell phenotype in specific regions of the culture as a function of hypoxia [228]. In addition to metabolic effects, this platform can be employed to characterize the effects of paracrine and non-soluble signaling between tumor and stromal cells [229,230].

Figure 4: Microfabricated models to study transport considerations.

Figure 4:

A. Schematic demonstrating transport considerations in the tumor microenvironment, including gradients of secreted factors, nutrients and oxygen, and elevated interstitial flow and pressure. Microfabricated in vitro models can isolate these aspects to determine their contribution to tumorigenesis. Image was created with BioRender.com. B. A TRACER model consisting of a sheet of cells rolled onto an oxygen impermeable membrane creates a naturally derived oxygen and nutrient gradient enabling spatial mapping of cell metabolism and phenotype. Reproduced with permission from Springer Nature [228]. C. An in vitro 3D culture model consisting of a Boyden chamber containing a cell-laden hydrogel was used to mimic the effects of interstitial flow in conjunction with chemokine signaling on tumor cell migration through a 3D matrix. Reproduced with permission from Elsevier [237]. D. A PDMS-based microfluidic vascularized model composed of an endothelial cell-coated channel (green), a central hydrogel channel, and a tumor cell channel (red) was used to investigate how endothelial barrier function influences tumor cell intravasation. Reproduced with permission from the National Academy of Sciences [244]. E. Endothelial cell-coated microfluidic channels with and without pericyte stabilization can also be formed directly within a collagen bulk preventing cell-contact with PDMS surfaces that may cause artefacts. Reproduced with permission from Springer Nature [252].

Similar to models that enable studies in the presence of soluble factor gradients, microfluidic platforms have been designed to precisely control fluid flow regimes and geometry in 3D cultures and investigate their effects on tumor cell behavior and malignancy. To study interstitial fluid flow, microfluidic devices often contain a 3D biomaterial region that is seeded with cells, where input channels or ports can then introduce flow across the biomaterial region through pressure differentials or gravity-driven flow [118,231233]. In one example, a PDMS microfluidic device consists of discrete cell-hydrogel compartments that are connected to microfluidic fluid channels, allowing for distinct spatial organization of different cell types and allowing for the investigation of interstitial flow on cellular communication [234]. Other examples include a modified Boyden chamber, in which cells embedded in a 3D hydrogel experience gravity driven-flow allowing for a relatively simple and reproducible alternative to investigate invasion due to interstitial flow (Figure 4C) [235237]. Studies using these devices revealed that interstitial flow not only directly modulates the behavior of stromal cells, but also the distribution of growth-factors through MMP-mediated release of ECM-bound molecules to synergistically enhance tumor cell invasion [119]. Devices to study the effect of interstitial pressure can be created utilizing micromolded hydrogels that lack an output port and lead to accumulation of pressure at an ECM/liquid interface [115,238]. Such models suggested that increased pressure enhances tumor cell invasion.

3.3.2. Engineered models of the vasculature

Although the above models have been crucial to understanding the individual contributions of transport considerations in the tumor microenvironment, they lack functional vasculature. Tissue-engineered microvascular models provide platforms to investigate tumor-endothelial cell interactions in the context of transport phenomena [239]. Several technologies utilize PDMS-based microfluidic devices consisting of three parallel channels where the center channel can be filled with a hydrogel component such as collagen or fibrin to allow cell-matrix interactions. Endothelial cells can then be introduced into the side channel(s) to form an endothelial barrier at the matrix interface, or within the hydrogel to self-assemble into a vascular network [240,241]. These devices have been used to study the influence of growth factors and fluid forces in angiogenesis, finding that VEGF gradients and flow direction can influence angiogenic sprouting [240,242,243]. Tumor and other stromal cells could also be introduced into the hydrogel or perfused through the vascular network. This approach allows for the study of tumor cell intra- and extravasation [244246] and has identified that macrophage-induced endothelial barrier impairment enhances tumor cell intravasation (Figure 4D) [244]. More recently, this design has been modified to incorporate a stromal region in between the tumor and endothelial regions, allowing for simultaneous investigation of cancer cell invasion into adjacent stroma and reciprocal tumor-vascular signaling under multiple ECM conditions [247,248]. One limitation of devices fabricated by soft-lithography is that cells may contact the PDMS surface, resulting in undesirable interactions at the device boundaries. Using a needle to form channels in a hydrogel for subsequent generation of a fully endothelialized lumen can help remedy this challenge [249,250]. However, as with the previous method, the architectures that can be obtained are limited to single tubes. Additionally, the size of endothelial lumens in these platforms is typically above a diameter of 100 μm due to device fabrication and stability constraints, whereas the average capillary diameter is an order of magnitude smaller [251]. By using a series of prefabricated molds or sacrificial scaffolds, more complex vascular architectures that include branched features reminiscent of physiological capillaries can be achieved (Figure 4E) [252254].

The vascular platforms described in this section are particularly well suited to investigating how vessel permeability, angiogenic sprouting, and cellular composition can affect the delivery of nutrients and growth factors [255,256]. In addition, reciprocal tumor-endothelial interactions could be studied in the context of paracrine signaling, drug testing, and tumor metabolism [36,257,258]. The nature of these in vitro vascular platforms facilitates dynamic imaging approaches and has allowed visualization of both angiogenic processes and intra- and extravasation kinetics [242,244,259]. However, endothelial cells cultured in vitro require specialized media with growth factor supplementation to survive and self-assemble into networks. To this end, recent advancements have been made in genetically modifying endothelial cells to transiently reactivate ETS variant transcription factor 2 (ETV2), allowing them to self-organize into adaptable, 3D lumenized vascular networks without the use of exogenous growth factors that could confound cell-cell signaling studies [260]. By integrating these systems with biochemical gradients and/or interstitial fluid flow/pressure, these engineered vasculature models will play a critical role in delineating the interplay between cell signaling and mass transport in the tumor microenvironment.

3.4. Next generation engineered tumor models

3.4.1. Organ/Body-on-a-chip models

Organ- and body-on-a-chip models recapitulate organ level functions or even systemic interactions between multiple organs, enabling more detailed analysis of cancer as a multistage and multi-organ process [261]. The majority of organ-on-a-chip models are manufactured in a similar manner as the microfluidic systems discussed in the previous section, but their design includes more specialized components, biomaterials, and cell populations. For example, organ-on-a-chip models have been developed to recapitulate the blood brain barrier, liver, and the intestine [262264], and have enabled studies of primary tumor growth in breast tissue, immune cell-mediated tumor cell migration, and tumor cell dormancy in liver [265267]. Importantly, organ-on-a-chip systems enable investigations into how the cellular, physical, and structural heterogeneity of specific target organs regulate processes relevant to metastasis [268]. For example, incorporating endothelial cells, bone marrow stem cells, and mineralized bone matrix into a microfluidic flow chamber to investigate bone-specific metastatic colonization has demonstrated that interstitial flow promotes the formation of stable vasculature and mediates metastatic potential within the skeleton (Figure 5A) [269]. Organ-on-a-chip systems can also reproduce the unique mechanical function and structural organization of lung tissue to study lung cancer and metastasis [270]. One such model is composed of two microchannels separated by a thin, porous, and flexible PDMS membrane that is seeded with alveolar epithelial cells and pulmonary microvascular endothelial cells on opposite sides and stretched using actuated vacuum chambers to reproduce the mechanical effects of breathing [270]. Indeed, this setup recapitulates central functions of lung tissue and revealed that cyclic mechanical strain involved in breathing increased therapeutic resistance and dormancy characteristics of tumor cells [271]. In both of these examples, organ-on-a-chip systems revealed how the specialized biophysical function and cellular heterogeneity of metastasis-prone tissues regulate organ-specific metastasis, insights that would not be possible with traditional in vitro or in vivo models.

Figure 5: Organ/body-on-a-chip systems and 3d printed models.

Figure 5:

A. Organ-on-a-chip models have been used to study bone-specific metastatic colonization by microfluidic integration of mineralized bone matrix, bone marrow stem cells, and endothelial cells. This device also enabled interstitial flow generation, revealing that flow increased cancer cell proliferation. Reproduced with permission from The National Academy of Sciences [269]. B. By integrating multiple organ-on-a-chip systems, a body-on-a-chip platform was used to measure anti-tumor efficacy and cardiotoxicity, demonstrating that the treatment responses of Ewing Sarcoma tumors and heart muscle to Linsitinib was closer to clinical trial results in this integrated system compared to treatment in isolated tissues. Reproduced with permission from The Royal Society of Chemistry [273]. C. Additive 3D bioprinting enables the fabrication of tissue scale constructs with complicated structures in conjunction with a temperature sensitive hydrogel support bath. The feasibility of this approach was demonstrated by printing, (i) a heart valve and (ii) a subregion of vascularized cardiac tissue. Reproduced with permission from The American Association for the Advancement of Science [282]. D. Negative-space bioprinting involves casting a material such as agarose around a 3D printed sacrificial template, which can generate complex architectures, for example, in vascular beds. Reproduced with permission from Springer Nature [287].

Body-on-a-chip platforms are even more complex and integrate cell types from multiple tissues or multiple organ-on-a-chip models to study how tissue-tissue interactions influence tumor-mediated systemic effects or anti-cancer drug toxicity [272]. Incorporating tumor cells, endothelial cells, and cardiomyocytes in microfluidic models, for example, permits simultaneous assessment of anti-tumor drug efficacy as well as cardiac safety [273,274]. Furthermore, integrating a bone Ewing sarcoma model and human cardiac tissue using microfluidics revealed that the drug Linsitinib had limited tumor response and reduced cardiotoxicity in the integrated system compared to either tissue type alone [273]. Importantly, these results were reminiscent of clinical trial results, suggesting that models integrating multiple tissue types could better predict clinical outcomes (Figure 5B).

In summary, organ- and body-on-a-chip systems represent the next generation of in vitro models that not only effectively capture the multiscale complexity of the tumor microenvironment, but also enable systems-level understanding of how cancer influences other organs and how these changes (or preconditions such as obesity or smoking), in turn, affect disease progression [272]. Combining these technologies with induced pluripotent stem cells (iPSCs) provides opportunities for experiments with human, organ-specific cells from a sustainable source [262,273275] that could help address challenges associated with primary cells. Despite these advantages, organ-on-a-chip systems can be more difficult to implement and typically have low throughput compared to traditional cell culture. The development of automated technologies to fluidically integrate and operate large numbers of organ-on-a-chip platforms simultaneously can help circumvent this limitation [276,277]. Furthermore, many body-on-a-chip technologies incorporate relatively simplistic organ compartments. Given their modular nature, however, they can be readily advanced by designing each compartment based on the parameters outlined above. For example, organ-on-a-chip systems incorporating patient-derived breast cancer organoids could provide an exciting platform for advancing drug discovery in personalized medicine settings [278]. Advancements such as these promise to reduce the use of animal models in preclinical testing of anti-cancer drugs.

3.4.2. 3D bioprinted models

While microfabrication approaches have enabled sophisticated in vitro platforms for studies of cancer, their fabrication requires access to microfabrication equipment. Additionally, current platforms are limited to a planar geometry and are based on plastics, glass, or PDMS, which do not mimic the mechanical and chemical interfaces cells encounter in vivo. Moreover, PDMS can leach free oligomers into culture media that have been shown to affect cell behavior [279]. Tumor models developed using 3D bioprinting technologies provide opportunities to address these limitations. These technologies are derived from traditional 3D printing, a form of additive manufacturing where a material is deposited in a specified pattern layer by layer to form a 3D object [280]. Whereas traditional 3D printing typically uses a plastic material as its “ink”, 3D bioprinting combines biomaterials and cells as “bioinks” in order to manufacture 3D biological constructs [280]. Compared to other models discussed in this review, the increased resolution and thickness in the z-direction confers an advantage over traditional microfluidics and organ-on-a-chip models, which are relatively thin and planar. Moreover, the ability to 3D print different biomaterials and cells in discrete locations drastically improves the level of control over architecture and spatial organization of cells and matrix to better represent the tumor microenvironment.

A variety of 3D bioprinting techniques exists to generate in vitro models. Additive printing is a common method, where a bioink consisting of matrix material and/or cells is forced through a nozzle and deposited in discrete layers to build up a 3D structure layer by layer. The bioink is either extruded as a continuous stream (extrusion-based) or deposited as droplets in close contact (inkjet-based) and solidified through thermally-induced polymerization or post-print by chemical cross-linking [281,282]. Initially, printing soft biomaterials in complex shapes required the use of stiff support structures, which can now be circumvented by using of a support bath. This support bath is composed of a temperature sensitive hydrogel that provides mechanical support for 3D printed in situ polymerizing matrix materials such as collagen, fibrin, or alginate at low temperatures [282,283], but releases the fully printed structure when heated to 37°C (Figure 5C). A similar method utilizing a hydrogel support bath allows printing of multicellular spheroids that self-assemble into high-cell density microtissues with spatially controlled cell ratios [284]. However, extrusion and inkjet methods are currently limited in their ability to print microscale structures due to nozzle size limitations. Light-based methods such as digital light processing (DLP) and multiphoton scanning can photo-crosslink biomaterials and generate scaffolds with spatial resolutions of less than 10 μm [285]. In these methods, ultraviolet (UV) light in conjunction with a photomask or point-scanning lasers are used to photopolymerize a structure within a bulk material. A drawback of light-based methods is that they are limited to photopolymerizable materials and are unable to directly print cells due to phototoxicity and DNA damage from exposure to UV light or lasers [280]. Additionally, achieving high resolution patterning often requires expensive and complex microscopy equipment with multiphoton capabilities. To this end, a method that incorporates natural and synthetic food dyes as photoabsorbers in a photopolymerizable hydrogel has enabled projection stereolithography to create intricate vascular architectures without the use of multiphoton scanning [286].

Another alternative to additive 3D bioprinting is printing sacrificial templates in what is known as negative-space printing. In contrast to additive printing, this technique involves casting a material around a 3D printed object and then removing the object physically or dissolving it, leaving an empty “printed” shape within a bulk material. The sacrificial template is typically printed from a material that can be dissolved at physiological temperatures (37°C) or in aqueous media, such as carbohydrates or Pluronic-F127 [280]. This method has been extensively used for creating engineered vasculature with complex architectures, where the hollow lumens that are left after template removal are seeded with endothelial cells (Figure 5D) [253,287289]. In comparison to additive bioprinting techniques, sacrificial scaffolds are limited in their material choice and cells can only be introduced after the printed template is removed, rather than being included in the printing material.

3D bioprinting holds great promise in advancing the field of tumor engineering. For example, 3D bioprinted models can recreate the irregular and tortuous architecture of tumor-associated vasculature enabling studies of the resulting consequences on mass transport in tumors [286,290]. Furthermore, the bioinks used in 3D printing could capture tumor specific heterogeneity, for example, by including multiple cell types, biochemical cues, and tumor-specific decellularized ECM in a spatially resolved manner to recapitulate the tumor-stroma interactions at the tumor/host interface [291293]. Despite their advantages 3D bioprinted tumor models are not widely adopted in the field yet due to a number of limitations inherent to new technologies, such as the lack of standardized printers and the need for optimization of bioinks and biomaterials used for printing [280]. Additionally, 3D bioprinting is currently restricted to materials that can be polymerized in situ such as collagen, alginate, gelatin, and photopolymerizable polymers. As the technology continues to develop, 3D bioprinting will enable widespread fabrication of reproducible and complex in vitro models.

4. Future directions and conclusions

Tumor engineering strategies have increased understanding of the dynamic steps underlying cancer development, progression, and therapy response, but many opportunities exist to further advance the field and translate findings into improved therapies (Figure 6). For example, integrating the described models with advanced imaging methods will enable monitoring cell behavior in 3D without introducing artefacts. Multiphoton or single-photon autofluorescence imaging techniques in particular allow dynamic imaging without the use of fixation, labels, or dyes, and has been applied for rapid, non-destructive imaging of patient-derived cancer organoids [294,295]. In addition, optical coherence tomography (OCT) enables label-free imaging of cancer spheroids embedded into fibrillar matrices based on temporal sampling of back-scattered coherent light to correlate tumor-induced matrix deformation and tumor invasion [135,296]. To obtain mechanistic insights into how microenvironmental factors influence intratumoral heterogeneity multi-omic and sequencing technologies could be applied. One particularly promising technique is deterministic barcoding in tissue for spatial omics sequencing (DBiT-seq), which allows spatial barcoding of molecules in tissues for simultaneous proteomics, imaging, and genomics [297]. In contrast to traditional sequencing methods, DBiT-seq retains spatial information to correlate molecular profiles with tissue context [298]. Finally, computational methods can complement in vitro tumor models, especially by extrapolating the observed phenomena to timescales relevant to tumor development in patients. For example, computational approaches can predict tumor growth, invasion, and treatment outcomes in response to metabolic heterogeneity, with timescales ranging from days to years [299]. Such in silico models can be further enhanced by integrating microenvironmental information including spatial organization of cells, cell deformation, and changes in chemical gradients [300]. However, iterative coupling between experiments and computational modeling is required to increase the relevance of both approaches.

Figure 6: Opportunities to improve engineered tumor models.

Figure 6:

The individual tumor models presented in this review vary in their complexity and span a range of engineering approaches to fabricate them. Integrating multiple of these different models and approaches within one integrated system represents an opportunity to develop the next generation of tumor models. Additional opportunities can be derived from integrating technologies from other disciplines such as imaging, advanced omics, and computational approaches as well as clinical/patient-oriented aspects such as using these models for cancer drug screening in precision medicine settings and improving scale-up manufacturing. Image was created with BioRender.com.

Given their ability to mimic aspects of cancer in a patient-specific manner, engineered tumor microenvironments have the potential to advance precision medicine applications. Indeed, incorporation of patient-derived or patient-specific materials such as organoids and patient-derived xenografts (PDXs) would enhance the translational value of engineered tumor models as they better capture the cellular heterogeneity and evolution of cancer seen in patients. For example, sequencing of organoid lines generated from advanced prostate cancer specimens revealed a landscape of mutations that were acquired during treatments and metastasis [144]. As these mutations could affect both cell-microenvironment interactions and therapy response, drug screening of such cells should be performed under conditions in which both parameters can be controlled independently. Engineered tumor microenvironments enable such studies and make it possible to more accurately predict therapy in the context of precision medicine [278]. Moving forward, integration of microfluidics, micropatterning and bioprinting technologies will create high throughput screening platforms that further advance personalized drug screening and ultimately improve patient prognosis.

In summary, cancer constitutes a multifaceted disease in which tumor cells’ interactions with the microenvironment are just as important as their molecular abnormalities. In vitro tumor models that provide controlled and physiologically relevant conditions to isolate the contribution of cellular and acellular microenvironmental factors to cancer progression continue to advance the field by defining the underlying mechanisms. By integrating them with advanced imaging, multi-omics, and computational approaches, engineered tumor models have the potential to yield unprecedented insights that will inform novel approaches to cancer therapy and precision medicine ultimately improving patient outcomes.

5. Acknowledgements

The work described was supported by the Center on the Physics of Cancer Metabolism through Award Number 1U54CA210184-01 from the National Cancer Institute, the Breast Cancer Coalition of Rochester, and the Cornell NanoScale Science & Technology Facility (CNF), a member of the National Nanotechnology Coordinated Infrastructure (NNCI) supported by the National Science Foundation (Grant NNCI-2025233).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

6. References

  • [1].Siegel RL, Miller KD, Fuchs HE, Jemal A, Cancer Statistics, 2021, CA. Cancer J. Clin 71 (2021) 7–33. 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
  • [2].Gupta GP, Massagué J, Cancer Metastasis: Building a Framework, Cell. 127 (2006) 679–695. 10.1016/j.cell.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • [3].Aguirre-Ghiso JA, Sosa MS, Emerging Topics on Disseminated Cancer Cell Dormancy and the Paradigm of Metastasis, Annu. Rev. Cancer Biol 2 (2018) 377–393. 10.1146/annurev-cancerbio. [DOI] [Google Scholar]
  • [4].Bissell MJ, Radisky DC, Rizki A, Weaver VM, Petersen OW, The organizing principle: Microenvironmental influences in the normal and malignant breast, Differentiation. 70 (2002) 537–546. 10.1046/j.1432-0436.2002.700907.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Paget S, THE DISTRIBUTION OF SECONDARY GROWTHS IN CANCER OF THE BREAST., Lancet. 133 (1889) 571–573. 10.1016/S0140-6736(00)49915-0. [DOI] [PubMed] [Google Scholar]
  • [6].Ribatti D, Mangialardi G, Vacca A, Stephen Paget and the ‘seed and soil’ theory of metastatic dissemination, Clin. Exp. Med 6 (2006) 145–149. 10.1007/s10238-006-0117-4. [DOI] [PubMed] [Google Scholar]
  • [7].Weigelt B, Ghajar CM, Bissell MJ, The need for complex 3D culture models to unravel novel pathways and identify accurate biomarkers in breast cancer, Adv. Drug Deliv. Rev 69–70 (2014) 42–51. 10.1016/j.addr.2014.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Bissell MJ, Hines WC, Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression, Nat. Med 17 (2011) 320–329. 10.1038/nm.2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Weaver VM, Petersen OW, Wang F, Larabell CA, Briand P, Damsky C, Bissell MJ, Reversion of the malignant phenotype of human breast cells in three- dimensional culture and in vivo by integrin blocking antibodies, J. Cell Biol 137 (1997) 231–245. 10.1083/jcb.137.1.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Hanahan D, Weinberg RA, Hallmarks of cancer: The next generation, Cell. 144 (2011) 646–674. 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • [11].Stylianopoulos T, Munn LL, Jain RK, Reengineering the Physical Microenvironment of Tumors to Improve Drug Delivery and Efficacy: From Mathematical Modeling to Bench to Bedside, Trends in Cancer. 4 (2018) 292–319. 10.1016/j.trecan.2018.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Correia AL, Bissell MJ, The tumor microenvironment is a dominant force in multidrug resistance, Drug Resist. Updat 15 (2012) 39–49. 10.1016/j.drup.2012.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Julien S, Merino-Trigo A, Lacroix L, Pocard M, Goéré D, Mariani P, Landron S, Bigot L, Nemati F, Dartigues P, Weiswald L-B, Lantuas D, Morgand L, Pham E, Gonin P, Dangles-Marie V, Job B, Dessen P, Bruno A, Pierré A, De Thé H, Soliman H, Nunes M, Lardier G, Calvet L, Demers B, Prévost G, Vrignaud P, Roman-Roman S, Duchamp O, Berthet C, Characterization of a Large Panel of Patient-Derived Tumor Xenografts Representing the Clinical Heterogeneity of Human Colorectal Cancer, Clin. Cancer Res 18 (2012) 5314–5328. 10.1158/1078-0432.CCR-12-0372. [DOI] [PubMed] [Google Scholar]
  • [14].Flier JS, Underhill LH, Dvorak HF, Tumors: Wounds That Do Not Heal, N. Engl. J. Med 315 (1986) 1650–1659. 10.1056/NEJM198612253152606. [DOI] [PubMed] [Google Scholar]
  • [15].Foster DS, Longaker MT, Jeffrey A, Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA, The evolving relationship of wound healing and tumor stroma Find the latest version : The evolving relationship of wound healing and tumor stroma, J. Clin. Invest 3 (2018) 1–17. 10.1172/jci.insight.99911. [DOI] [Google Scholar]
  • [16].Kalluri R, Zeisberg M, Fibroblasts in cancer, Nat. Rev. Cancer 6 (2006) 392–401. 10.1038/nrc1877. [DOI] [PubMed] [Google Scholar]
  • [17].Charrier EE, Pogoda K, Li R, Park CY, Fredberg JJ, Janmey PA, A novel method to make viscoelastic polyacrylamide gels for cell culture and traction force microscopy, APL Bioeng. 4 (2020). 10.1063/5.0002750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, Hynes RO, Jain RK, Janowitz T, Jorgensen C, Kimmelman AC, Kolonin MG, Maki RG, Powers RS, Puré E, Ramirez DC, Scherz-Shouval R, Sherman MH, Stewart S, Tlsty TD, Tuveson DA, Watt FM, Weaver V, Weeraratna AT, Werb Z, A framework for advancing our understanding of cancer-associated fibroblasts, Nat. Rev. Cancer 20 (2020) 174–186. 10.1038/s41568-019-0238-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Chen WJ, Ho CC, Chang YL, Chen HY, Lin CA, Ling TY, Yu SL, Yuan SS, Louisa Chen YJ, Lin CY, Pan SH, Elizabeth Chou HY, Chang GC, Chu WC, Lee YM, Lee JY, Lee PJ, Li KC, Chen HW, Yang PC, Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling, Nat. Commun 5 (2014). 10.1038/ncomms4472. [DOI] [PubMed] [Google Scholar]
  • [20].Clapéron A, Mergey M, Aoudjehane L, Ho-Bouldoires THN, Wendum D, Prignon A, Merabtene F, Firrincieli D, Desbois-Mouthon C, Scatton O, Conti F, Housset C, Fouassier L, Hepatic myofibroblasts promote the progression of human cholangiocarcinoma through activation of epidermal growth factor receptor, Hepatology. 58 (2013) 2001–2011. 10.1002/hep.26585. [DOI] [PubMed] [Google Scholar]
  • [21].Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA, Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion, Cell. 121 (2005) 335–348. 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
  • [22].Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L, Karin M, IL-6 and Stat3 Are Required for Survival of Intestinal Epithelial Cells and Development of Colitis-Associated Cancer, Cancer Cell. 15 (2009) 103–113. 10.1016/j.ccr.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Lu P, Weaver VM, Werb Z, The extracellular matrix: A dynamic niche in cancer progression, J. Cell Biol 196 (2012) 395–406. 10.1083/jcb.201102147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Chabottaux V, Noel A, Breast cancer progression: Insights into multifaceted matrix metalloproteinases, Clin. Exp. Metastasis 24 (2007) 647–656. 10.1007/s10585-007-9113-7. [DOI] [PubMed] [Google Scholar]
  • [25].Monteran L, Erez N, The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment, Front. Immunol 10 (2019) 1–15. 10.3389/fimmu.2019.01835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].O’Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI, LeBleu VS, Dewar R, Rocha RM, Brentani RR, Resnick MB, Neilson EG, Zeisberg M, Kalluri R, VEGF-A and Tenascin-C produced by S100A4 + stromal cells are important for metastatic colonization, Proc. Natl. Acad. Sci. U. S. A 108 (2011) 16002–16007. 10.1073/pnas.1109493108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Loeffler M, Niethammer AG, Ralph A, Loeffler M, Krüger JA, Niethammer AG, Reisfeld RA, Targeting tumor-associated fibroblasts improves cancer chemotherapy by increasing intratumoral drug uptake Find the latest version : Targeting tumor-associated fibroblasts improves cancer chemotherapy by increasing intratumoral drug uptake, (2009). 10.1172/JCI26532.modulation. [DOI] [Google Scholar]
  • [28].Yasunaga M, Manabe S, Tarin D, Matsumura Y, Cancer-Stroma Targeting Therapy by Cytotoxic Immunoconjugate Bound to the Collagen 4 Network in the Tumor Tissue, (2011) 1776–1783. 10.1021/bc200158j. [DOI] [PubMed] [Google Scholar]
  • [29].Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, Mcintyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Rückert F, Grützmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Inhibition of Hedgehog Signaling, (2009) 1457–1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, DeJesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R, Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival, Cancer Cell. 25 (2014) 719–734. 10.1016/j.ccr.2014.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Valastyan S, Weinberg RA, Tumor metastasis: Molecular insights and evolving paradigms, Cell. 147 (2011) 275–292. 10.1016/j.cell.2011.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Carmeliet P, Jain RK, Angiogenesis in cancer and other diseases, Nature. 407 (2000) 249–257. 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
  • [33].Lugano R, Ramachandran M, Dimberg A, Tumor angiogenesis: causes, consequences, challenges and opportunities, Cell. Mol. Life Sci 77 (2020) 1745–1770. 10.1007/s00018-019-03351-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Carmeliet P, Jain RK, Molecular mechanisms and clinical applications of angiogenesis, Nature. 473 (2011) 298–307. 10.1038/nature10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Balkwill FR, Capasso M, Hagemann T, The tumor microenvironment at a glance, J. Cell Sci 125 (2012) 5591–5596. 10.1242/jcs.116392. [DOI] [PubMed] [Google Scholar]
  • [36].Kutys ML, Polacheck WJ, Welch MK, Gagnon KA, Koorman T, Kim S, Li L, McClatchey AI, Chen CS, Uncovering mutation-specific morphogenic phenotypes and paracrine-mediated vessel dysfunction in a biomimetic vascularized mammary duct platform, Nat. Commun 11 (2020). 10.1038/s41467-020-17102-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Martin JD, Seano G, Jain RK, Normalizing Function of Tumor Vessels: Progress, Opportunities, and Challenges, Annu. Rev. Physiol 81 (2019) 505–534. 10.1146/annurev-physiol-020518-114700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Munn LL, Aberrant vascular architecture in tumors and its importance in drug-based therapies, Drug Discov. Today 8 (2003) 396–403. 10.1016/S1359-6446(03)02686-2. [DOI] [PubMed] [Google Scholar]
  • [39].Lyssiotis CA, Kimmelman AC, Metabolic Interactions in the Tumor Microenvironment, Trends Cell Biol. 27 (2017) 863–875. 10.1016/j.tcb.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Cao Z, Sen Ding B-S, Guo P, Lee SB, Butler JM, Casey SC, Simons M, Tam W, Felsher DW, Shido K, Rafii A, Scandura JM, Rafii S, Angiocrine Factors Deployed by Tumor Vascular Niche Induce B Cell Lymphoma Invasiveness and Chemoresistance, Cancer Cell. 25 (2014) 350–365. 10.1016/j.ccr.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Butler JM, Kobayashi H, Rafii S, Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors, Nat. Rev. Cancer 10 (2010) 138–146. 10.1038/nrc2791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Cao Z, Scandura JM, Inghirami GG, Shido K, Sen Ding B-S, Rafii S, Molecular Checkpoint Decisions Made by Subverted Vascular Niche Transform Indolent Tumor Cells into Chemoresistant Cancer Stem Cells, Cancer Cell. 31 (2017) 110–126. 10.1016/j.ccell.2016.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Infanger DW, Cho Y, Lopez BS, Mohanan S, Liu SC, Gursel D, Boockvar JA, Fischbach C, Glioblastoma Stem Cells Are Regulated by Interleukin-8 Signaling in a Tumoral Perivascular Niche, Cancer Res. 73 (2013) 7079–7089. 10.1158/0008-5472.CAN-13-1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].McCoy MG, Nyanyo D, Hung CK, Goerger JP, Zipfel WR, Williams RM, Nishimura N, Fischbach C, Endothelial cells promote 3D invasion of GBM by IL-8-dependent induction of cancer stem cell properties, Sci. Rep 9 (2019) 1–14. 10.1038/s41598-019-45535-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, Oh EY, Gaber MW, Finklestein D, Allen M, Frank A, Bayazitov IT, Zakharenko SS, Gajjar A, Davidoff A, Gilbertson RJ, A Perivascular Niche for Brain Tumor Stem Cells, Cancer Cell. 11 (2007) 69–82. 10.1016/j.ccr.2006.11.020. [DOI] [PubMed] [Google Scholar]
  • [46].Yan G-N, Yang L, Lv Y, Shi Y, Shen L-L, Yao X-H, Guo Q-N, Zhang P, Cui Y-H, Zhang X, Bian X-W, Guo D-Y, Endothelial cells promote stem-like phenotype of glioma cells, J. Pathol 234 (2014) 11–22. 10.1002/path.4349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Pietras A, Katz AM, Ekström EJ, Wee B, Halliday JJ, Pitter KL, Werbeck JL, Amankulor NM, Huse JT, Holland EC, Osteopontin-CD44 Signaling in the Glioma Perivascular Niche Enhances Cancer Stem Cell Phenotypes and Promotes Aggressive Tumor Growth, Cell Stem Cell. 14 (2014) 357–369. 10.1016/j.stem.2014.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Lu J, Ye X, Fan F, Xia L, Bhattacharya R, Bellister S, Tozzi F, Sceusi E, Zhou Y, Tachibana I, Maru DM, Hawke DH, Rak J, Mani SA, Zweidler-McKay P, Ellis LM, Endothelial Cells Promote the Colorectal Cancer Stem Cell Phenotype through a Soluble Form of Jagged-1, Cancer Cell. 23 (2013) 171–185. 10.1016/j.ccr.2012.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].McCoy MG, Nyanyo D, Hung CK, Goerger JP, Zipfel WR, Williams RM, Nishimura N, Fischbach C, Endothelial cells promote 3D invasion of GBM by IL-8-dependent induction of cancer stem cell properties, Sci. Rep 9 (2019) 1–14. 10.1038/s41598-019-45535-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Carlson P, Dasgupta A, Grzelak CA, Kim J, Barrett A, Coleman IM, Shor RE, Goddard ET, Dai J, Schweitzer EM, Lim AR, Crist SB, Cheresh DA, Nelson PS, Hansen KC, Ghajar CM, Targeting the perivascular niche sensitizes disseminated tumour cells to chemotherapy, Nat. Cell Biol 21 (2019) 238–250. 10.1038/s41556-018-0267-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, Almeida D, Koller A, Hajjar KA, Stainier DYR, Chen EI, Lyden D, Bissell MJ, The perivascular niche regulates breast tumour dormancy, Nat. Cell Biol 15 (2013) 807–817. 10.1038/ncb2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Greten FR, Grivennikov SI, Inflammation and Cancer: Triggers, Mechanisms, and Consequences, Immunity. 51 (2019) 27–41. 10.1016/j.immuni.2019.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Lewis CE, Harney AS, Pollard JW, The Multifaceted Role of Perivascular Macrophages in Tumors, Cancer Cell. 30 (2016) 18–25. 10.1016/j.ccell.2016.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Pathria P, Louis TL, Varner JA, Targeting Tumor-Associated Macrophages in Cancer, Trends Immunol. 40 (2019) 310–327. 10.1016/j.it.2019.02.003. [DOI] [PubMed] [Google Scholar]
  • [55].Linde N, Casanova-Acebes M, Sosa MS, Mortha A, Rahman A, Farias E, Harper K, Tardio E, Reyes Torres I, Jones J, Condeelis J, Merad M, Aguirre-Ghiso JA, Macrophages orchestrate breast cancer early dissemination and metastasis, Nat. Commun 9 (2018) 21. 10.1038/s41467-017-02481-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Condeelis J, Pollard JW, Macrophages: Obligate Partners for Tumor Cell Migration, Invasion, and Metastasis, Cell. 124 (2006) 263–266. 10.1016/j.cell.2006.01.007. [DOI] [PubMed] [Google Scholar]
  • [57].Arwert EN, Harney AS, Entenberg D, Wang Y, Sahai E, Pollard JW, Condeelis JS, A Unidirectional Transition from Migratory to Perivascular Macrophage Is Required for Tumor Cell Intravasation, Cell Rep. 23 (2018) 1239–1248. 10.1016/j.celrep.2018.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Noy R, Pollard JW, Tumor-Associated Macrophages: From Mechanisms to Therapy, Immunity. 41 (2014) 49–61. 10.1016/j.immuni.2014.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Springer NL, Iyengar NM, Bareja R, Verma A, Jochelson MS, Giri DD, Zhou XK, Elemento O, Dannenberg AJ, Fischbach C, Obesity-Associated Extracellular Matrix Remodeling Promotes a Macrophage Phenotype Similar to Tumor-Associated Macrophages, Am. J. Pathol 189 (2019) 2019–2035. 10.1016/j.ajpath.2019.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Gajewski TF, Schreiber H, Fu Y-X, Innate and adaptive immune cells in the tumor microenvironment, Nat. Immunol 14 (2013) 1014–1022. 10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N, Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade, Proc. Natl. Acad. Sci. U. S. A 99 (2002) 12293–12297. 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K, Katsaros D, O’Brien-Jenkins A, Gimotty PA, Coukos G, Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy, Nat. Med 14 (2008) 28–36. 10.1038/nm1699. [DOI] [PubMed] [Google Scholar]
  • [63].Liu T, Han C, Wang S, Fang P, Ma Z, Xu L, Yin R, Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy, J. Hematol. Oncol 12 (2019) 1–15. 10.1186/s13045-019-0770-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z, Concepts of extracellular matrix remodelling in tumour progression and metastasis, Nat. Commun 11 (2020) 1–19. 10.1038/s41467-020-18794-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Kalluri R, Basement membranes: Structure, assembly and role in tumour angiogenesis, Nat. Rev. Cancer 3 (2003) 422–433. 10.1038/nrc1094. [DOI] [PubMed] [Google Scholar]
  • [66].Chang J, Chaudhuri O, Beyond proteases: Basement membrane mechanics and cancer invasion, J. Cell Biol 218 (2019) 2456–2469. 10.1083/JCB.201903066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Naba A, Clauser KR, Hoersch S, Liu H, Carr SA, Hynes RO, The Matrisome: In Silico Definition and In Vivo Characterization by Proteomics of Normal and Tumor Extracellular Matrices, 2012. 10.1074/mcp.M111.014647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Zhang K, Corsa CA, Ponik SM, Prior JL, Piwnica-Worms D, Eliceiri KW, Keely PJ, Longmore GD, The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis, Nat. Cell Biol 15 (2013) 677–687. 10.1038/ncb2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM, Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling, Cell. 139 (2009) 891–906. 10.1016/j.cell.2009.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Wang K, Andresen Eguiluz RC, Wu F, Seo BR, Fischbach C, Gourdon D, Stiffening and unfolding of early deposited-fibronectin increase proangiogenic factor secretion by breast cancer-associated stromal cells, Biomaterials. 54 (2015) 63–71. 10.1016/j.biomaterials.2015.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Attieh Y, Clark AG, Grass C, Richon S, Pocard M, Mariani P, Elkhatib N, Betz T, Gurchenkov B, Vignjevic DM, Cancer-associated fibroblasts lead tumor invasion through integrin-β3–dependent fibronectin assembly, J. Cell Biol 216 (2017) 3509–3520. 10.1083/jcb.201702033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Bourguignon LYW, Peyrollier K, Xia W, Gilad E, Hyaluronan-CD44 interaction activates stem cell marker Nanog, Stat-3-mediated MDR1 gene expression, and ankyrin-regulated multidrug efflux in breast and ovarian tumor cells, J. Biol. Chem 283 (2008) 17635–17651. 10.1074/jbc.M800109200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, Lolkema MP, Jiang P, Kultti A, Thompson CB, Maneval DC, Jodrell DI, Frost GI, Shepard HM, Skepper JN, Tuveson DA, Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer, Gut. 62 (2013) 112–120. 10.1136/gutjnl-2012-302529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Auvinen P, Rilla K, Tumelius R, Tammi M, Sironen R, Soini Y, Kosma VM, Mannermaa A, Viikari J, Tammi R, Hyaluronan synthases (HAS1–3) in stromal and malignant cells correlate with breast cancer grade and predict patient survival, Breast Cancer Res. Treat 143 (2014) 277–286. 10.1007/s10549-013-2804-7. [DOI] [PubMed] [Google Scholar]
  • [75].Yayon A, Klagsbrun M, Esko JD, Leder P, Ornitz DM, Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor, Cell. 64 (1991) 841–848. 10.1016/0092-8674(91)90512-W. [DOI] [PubMed] [Google Scholar]
  • [76].Gitay-gorens H, Sokers S, Vlodavskys I, Neufeldsli G, The Binding of Vascular Endothelial Growth Factor Its to Receptors Is Dependent on Cell Surface-associated Heparin-like Molecules *, (1992). 10.1016/S0021-9258(18)42666-X. [DOI] [PubMed] [Google Scholar]
  • [77].Mitsi M, Forsten-Williams K, Gopalakrishnan M, Nugent MA, A catalytic role of heparin within the extracellular matrix, J. Biol. Chem 283 (2008) 34796–34807. 10.1074/jbc.M806692200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Frantz C, Stewart KM, Weaver VM, The extracellular matrix at a glance, J. Cell Sci 123 (2010) 4195–4200. 10.1242/jcs.023820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ, Collagen reorganization at the tumor-stromal interface facilitates local invasion, BMC Med. 4 (2006) 1–15. 10.1186/1741-7015-4-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Falzon G, Pearson S, Murison R, Analysis of collagen fibre shape changes in breast cancer, Phys. Med. Biol 53 (2008) 6641–6652. 10.1088/0031-9155/53/23/001. [DOI] [PubMed] [Google Scholar]
  • [81].Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, Friedl A, Keely PJ, Aligned collagen is a prognostic signature for survival in human breast carcinoma, Am. J. Pathol 178 (2011) 1221–1232. 10.1016/j.ajpath.2010.11.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Seo BR, Chen X, Ling L, Song YH, Shimpi AA, Choi S, Gonzalez J, Sapudom J, Wang K, Eguiluz RCA, Gourdon D, Shenoy VB, Fischbach C, Collagen microarchitecture mechanically controls myofibroblast differentiation, Proc. Natl. Acad. Sci. U. S. A 117 (2020). 10.1073/pnas.1919394117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Sugimoto K, Ishibashi T, Sawamura T, Inoue N, Kamioka M, Uekita H, Ohkawara H, Sakamoto T, Sakamoto N, Okamoto Y, Takuwa Y, Kakino A, Fujita Y, Tanaka T, Teramoto T, Maruyama Y, Takeishi Y, LOX-1-MT1-MMP axis is crucial for RhoA and Rac1 activation induced by oxidized low-density lipoprotein in endothelial cells, Cardiovasc. Res 84 (2009) 127–136. 10.1093/cvr/cvp177. [DOI] [PubMed] [Google Scholar]
  • [84].Chaudhuri O, Cooper-White J, Janmey PA, Mooney DJ, Shenoy VB, Effects of extracellular matrix viscoelasticity on cellular behaviour, Nature. 584 (2020) 535–546. 10.1038/s41586-020-2612-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Hammer DA, Weaver VM, Tensional homeostasis and the malignant phenotype, Cancer Cell. 8 (2005) 241–254. 10.1016/j.ccr.2005.08.010. [DOI] [PubMed] [Google Scholar]
  • [86].Wirtz P, Konstantopoulos D, Searson K, Mechanical Forces in Metastasis, Biomol. Eng 11 (2012) 512–522. 10.1038/nrc3080.The. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ, Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells, Proc. Natl. Acad. Sci. U. S. A 89 (1992) 9064–9068. 10.1073/pnas.89.19.9064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Chaudhuri O, Koshy ST, Branco Da Cunha C, Shin JW, Verbeke CS, Allison KH, Mooney DJ, Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium, Nat. Mater 13 (2014) 970–978. 10.1038/nmat4009. [DOI] [PubMed] [Google Scholar]
  • [89].Kubow KE, Vukmirovic R, Zhe L, Klotzsch E, Smith ML, Gourdon D, Luna S, Vogel V, Mechanical forces regulate the interactions of fibronectin and collagen i in extracellular matrix, Nat. Commun 6 (2015). 10.1038/ncomms9026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Smith ML, Gourdon D, Little WC, Kubow KE, Eguiluz RA, Luna-morris S, Vogel V, Force-Induced Unfolding of Fibronectin in the Extracellular Matrix of Living Cells, 5 (2007). 10.1371/journal.pbio.0050268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA, Myofibroblasts and mechano: Regulation of connective tissue remodelling, Nat. Rev. Mol. Cell Biol 3 (2002) 349–363. 10.1038/nrm809. [DOI] [PubMed] [Google Scholar]
  • [92].DelNero P, Hopkins BD, Cantley LC, Fischbach C, Cancer metabolism gets physical, Sci. Transl. Med 10 (2018) 1–8. 10.1126/scitranslmed.aaq1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lyssiotis CA, Kimmelman AC, Metabolic Interactions in the Tumor Microenvironment, Trends Cell Biol. 27 (2017) 863–875. 10.1016/j.tcb.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Nia HT, Munn LL, Jain RK, Physical traits of cancer, Science (80-. ). 370 (2020) eaaz0868. 10.1126/science.aaz0868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Schito L, Semenza GL, Hypoxia-Inducible Factors: Master Regulators of Cancer Progression, Trends in Cancer. 2 (2016) 758–770. 10.1016/j.trecan.2016.10.016. [DOI] [PubMed] [Google Scholar]
  • [96].Nakazawa MS, Keith B, Simon MC, Oxygen availability and metabolic adaptations, Nat. Rev. Cancer 16 (2016) 663–673. 10.1038/nrc.2016.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Wong CC-L, Gilkes DM, Zhang H, Chen J, Wei H, Chaturvedi P, Fraley SI, Wong C-M, Khoo U-S, Ng IO-L, Wirtz D, Semenza GL, Hypoxia-inducible factor 1 is a master regulator of breast cancer metastatic niche formation, Proc. Natl. Acad. Sci 108 (2011) 16369–16374. 10.1073/pnas.1113483108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Fluegen G, Avivar-Valderas A, Wang Y, Padgen MR, Williams JK, Nobre AR, Calvo V, Cheung JF, Bravo-Cordero JJ, Entenberg D, Castracane J, Verkhusha V, Keely PJ, Condeelis J, Aguirre-Ghiso JA, Phenotypic heterogeneity of disseminated tumour cells is preset by primary tumour hypoxic microenvironments, Nat. Cell Biol 19 (2017) 120–132. 10.1038/ncb3465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Pang M-F, Siedlik MJ, Han S, Stallings-Mann M, Radisky DC, Nelson CM, Tissue Stiffness and Hypoxia Modulate the Integrin-Linked Kinase ILK to Control Breast Cancer Stem-like Cells, Cancer Res. 76 (2016) 5277–5287. 10.1158/0008-5472.CAN-16-0579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Ju JA, Godet I, Ye IC, Byun J, Jayatilaka H, Lee SJ, Xiang L, Samanta D, Lee MH, Wu P-H, Wirtz D, Semenza GL, Gilkes DM, Hypoxia Selectively Enhances Integrin α 5 β 1 Receptor Expression in Breast Cancer to Promote Metastasis, Mol. Cancer Res 15 (2017) 723–734. 10.1158/1541-7786.MCR-16-0338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Gilkes DM, Bajpai S, Chaturvedi P, Wirtz D, Semenza GL, Hypoxia-inducible Factor 1 (HIF-1) Promotes Extracellular Matrix Remodeling under Hypoxic Conditions by Inducing P4HA1, P4HA2, and PLOD2 Expression in Fibroblasts*, J. Biol. Chem 288 (2013) 10819–10829. 10.1074/jbc.M112.442939. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [102].Semenza GL, Targeting HIF-1 for cancer therapy, Nat. Rev. Cancer 3 (2003) 721–732. 10.1038/nrc1187. [DOI] [PubMed] [Google Scholar]
  • [103].Zhang Q, Yan Q, Yang H, Wei W, Oxygen sensing and adaptability won the 2019 Nobel Prize in Physiology or medicine, Genes Dis. 6 (2019) 328–332. 10.1016/j.gendis.2019.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Keith B, Johnson RS, Simon MC, HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression, Nat. Rev. Cancer 12 (2012) 9–22. 10.1038/nrc3183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Lee JW, Ko J, Ju C, Eltzschig HK, Hypoxia signaling in human diseases and therapeutic targets, Exp. Mol. Med 51 (2019) 1–13. 10.1038/s12276-019-0235-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].DeBerardinis RJ, Chandel NS, Fundamentals of cancer metabolism, Sci. Adv 2 (2016) e1600200. 10.1126/sciadv.1600200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].García-Cañaveras JC, Chen L, Rabinowitz JD, The Tumor Metabolic Microenvironment: Lessons from Lactate, Cancer Res. 79 (2019) 3155–3162. 10.1158/0008-5472.CAN-18-3726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Estrella V, Chen T, Lloyd M, Wojtkowiak J, Cornnell HH, Ibrahim-Hashim A, Bailey K, Balagurunathan Y, Rothberg JM, Sloane BF, Johnson J, Gatenby RA, Gillies RJ, Acidity Generated by the Tumor Microenvironment Drives Local Invasion, Cancer Res. 73 (2013) 1524–1535. 10.1158/0008-5472.CAN-12-2796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Sonveaux P, Végran F, Schroeder T, Wergin MC, Verrax J, Rabbani ZN, De Saedeleer CJ, Kennedy KM, Diepart C, Jordan BF, Kelley MJ, Gallez B, Wahl ML, Feron O, Dewhirst MW, Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice, J. Clin. Invest 118 (2008) 3930–3942. 10.1172/JCI36843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Peppicelli S, Bianchini F, Toti A, Laurenzana A, Fibbi G, Calorini L, Extracellular acidity strengthens mesenchymal stem cells to promote melanoma progression, Cell Cycle. 14 (2015) 3088–3100. 10.1080/15384101.2015.1078032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfør K, Rofstad EK, Mueller-Klieser W, High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers, Cancer Res. 60 (2000) 916–921. [PubMed] [Google Scholar]
  • [112].Luo M, Brooks M, Wicha MS, Asparagine and Glutamine: Co-conspirators Fueling Metastasis, Cell Metab. 27 (2018) 947–949. 10.1016/j.cmet.2018.04.012. [DOI] [PubMed] [Google Scholar]
  • [113].Padera TP, Meijer EFJ, Munn LL, The Lymphatic System in Disease Processes and Cancer Progression, Annu. Rev. Biomed. Eng 18 (2016) 125–158. 10.1146/annurev-bioeng-112315-031200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Wiig H, Swartz MA, Interstitial Fluid and Lymph Formation and Transport: Physiological Regulation and Roles in Inflammation and Cancer, Physiol. Rev 92 (2012) 1005–1060. 10.1152/physrev.00037.2011. [DOI] [PubMed] [Google Scholar]
  • [115].Piotrowski-Daspit AS, Tien J, Nelson CM, Interstitial fluid pressure regulates collective invasion in engineered human breast tumors via Snail, vimentin, and E-cadherin, Integr. Biol 8 (2016) 319–331. 10.1039/c5ib00282f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Kingsmore KM, Logsdon DK, Floyd DH, Peirce SM, Purow BW, Munson JM, Interstitial flow differentially increases patient-derived glioblastoma stem cell invasion via CXCR4, CXCL12, and CD44-mediated mechanisms, Integr. Biol 8 (2016) 1246–1260. 10.1039/c6ib00167j. [DOI] [PubMed] [Google Scholar]
  • [117].Pisano M, Triacca V, Barbee KA, Swartz MA, An in vitro model of the tumor–lymphatic microenvironment with simultaneous transendothelial and luminal flows reveals mechanisms of flow enhanced invasion, Integr. Biol 7 (2015) 525–533. 10.1039/C5IB00085H. [DOI] [PubMed] [Google Scholar]
  • [118].Polacheck WJ, Charest JL, Kamm RD, Interstitial flow influences direction of tumor cell migration through competing mechanisms, Proc. Natl. Acad. Sci 108 (2011) 11115–11120. 10.1073/pnas.1103581108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Shieh AC, Rozansky HA, Hinz B, Swartz MA, Tumor Cell Invasion Is Promoted by Interstitial Flow-Induced Matrix Priming by Stromal Fibroblasts, Cancer Res. 71 (2011) 790–800. 10.1158/0008-5472.CAN-10-1513. [DOI] [PubMed] [Google Scholar]
  • [120].Li R, Serrano JC, Xing H, Lee TA, Azizgolshani H, Zaman M, Kamm RD, Interstitial flow promotes macrophage polarization toward an M2 phenotype, Mol. Biol. Cell 29 (2018) 1927–1940. 10.1091/mbc.E18-03-0164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].and Lapidot JED, Tsvee, Sirard Christian, Vormoor Josef, Murdoch Barbara, Hoang Trang, Caceres-Cortes Julio, Minden Mark, Paterson Bruce, Caligiuri Michael A., A cell initiating human acute myeloid leukaemia after transplantation into SCID mice No Title, Nature. 367 (1994) 645–648. [DOI] [PubMed] [Google Scholar]
  • [122].Zanoni M, Piccinini F, Arienti C, Zamagni A, Santi S, Polico R, Bevilacqua A, Tesei A, 3D tumor spheroid models for in vitro therapeutic screening: A systematic approach to enhance the biological relevance of data ob[1] M. Zanoni, F. Piccinini, C. Arienti, A. Zamagni, S. Santi, R. Polico, A. Bevilacqua, A. Tesei, Sci. Rep 2016, 6, 1.tained, Sci. Rep.6 (2016) 1–11. 10.1038/srep19103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Hinohara K, Polyak K, Intratumoral Heterogeneity: More Than Just Mutations, Trends Cell Biol. 29 (2019) 569–579. 10.1016/j.tcb.2019.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Duval K, Grover H, Han LH, Mou Y, Pegoraro AF, Fredberg J, Chen Z, Modeling physiological events in 2D vs. 3D cell culture, Physiology. 32 (2017) 266–277. 10.1152/physiol.00036.2016. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
  • [125].Kunz-Schughart LA, Freyer JP, Hofstaedter F, Ebner R, The use of 3-D cultures for high-throughput screening: The multicellular spheroid model, J. Biomol. Screen 9 (2004) 273–285. 10.1177/1087057104265040. [DOI] [PubMed] [Google Scholar]
  • [126].Li Y, Kumacheva E, Hydrogel microenvironments for cancer spheroid growth and drug screening, Sci. Adv 4 (2018) 1–11. 10.1126/sciadv.aas8998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Foty R, A simple hanging drop cell culture protocol for generation of 3D spheroids, J. Vis. Exp 51 (2011). 10.3791/2720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Mehta G, Hsiao AY, Ingram M, Luker GD, Takayama S, Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy, J. Control. Release 164 (2012) 192–204. 10.1016/j.jconrel.2012.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Ingram M, Techy GB, Saroufeem R, Yazan O, Narayan KS, Goodwin TJ, Spaulding GF, Three-dimensional growth patterns of various human tumor cell lines in simulated microgravity of a nasa bioreactor, Vitr. Cell. Dev. Biol. - Anim 33 (1997) 459–466. 10.1007/s11626-997-0064-8. [DOI] [PubMed] [Google Scholar]
  • [130].Hsiao AY, Torisawa Y, Tung Y, Sud S, Taichman RS, Pienta KJ, Takayama S, Microfluidic system for formation of PC-3 prostate cancer co-culture spheroids, Biomaterials. 30 (2009) 3020–3027. 10.1016/j.biomaterials.2009.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Bin Lee Y, Kim EM, Byun H, kwan Chang H, Jeong K, Aman ZM, Choi YS, Park J, Shin H, Engineering spheroids potentiating cell-cell and cell-ECM interactions by self-assembly of stem cell microlayer, Biomaterials. 165 (2018) 105–120. 10.1016/j.biomaterials.2018.02.049. [DOI] [PubMed] [Google Scholar]
  • [132].Fukuda J, Khademhosseini A, Yeo Y, Yang X, Yeh J, Eng G, Blumling J, Wang C, Kohane DS, Langer R, Micromolding of photocrosslinkable chitosan hydrogel for spheroid microarray and co-cultures, 27 (2006) 5259–5267. 10.1016/j.biomaterials.2006.05.044. [DOI] [PubMed] [Google Scholar]
  • [133].Maguire SL, Peck B, Wai PT, Campbell J, Barker H, Gulati A, Daley F, Vyse S, Huang P, Lord CJ, Farnie G, Brennan K, Natrajan R, Three-dimensional modelling identifies novel genetic dependencies associated with breast cancer progression in the isogenic MCF10 model, J. Pathol 240 (2016) 315–328. 10.1002/path.4778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Horman SR OA, To J, Lamb J, Zoll JH, Leonetti N, Tu B, Moran R, Newlin R, Walker JR, Functional profiling of microtumors to identify cancer associated fibroblast-derived drug targets, Oncotarget. 8 (2017) 99913–99930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Ling L, Mulligan JA, Ouyang Y, Shimpi AA, Williams RM, Beeghly GF, Hopkins BD, Spector JA, Adie SG, Fischbach C, Obesity-Associated Adipose Stromal Cells Promote Breast Cancer Invasion through Direct Cell Contact and ECM Remodeling, Adv. Funct. Mater 1910650 (2020) 1–13. 10.1002/adfm.201910650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Rebelo SP, Pinto C, Martins TR, Harrer N, Estrada MF, Loza-Alvarez P, Cabeçadas J, Alves PM, Gualda EJ, Sommergruber W, Brito C, 3D-3-culture: A tool to unveil macrophage plasticity in the tumour microenvironment, Biomaterials. 163 (2018) 185–197. 10.1016/j.biomaterials.2018.02.030. [DOI] [PubMed] [Google Scholar]
  • [137].Drost J, Clevers H, Organoids in cancer research, Nat. Rev. Cancer 18 (2018) 407–418. 10.1038/s41568-018-0007-6. [DOI] [PubMed] [Google Scholar]
  • [138].Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H, A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity, Cell. 172 (2018) 373–386.e10. 10.1016/j.cell.2017.11.010. [DOI] [PubMed] [Google Scholar]
  • [139].Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, Chio IIC, Il Hwang C, Tiriac H, Baker LA, Engle DD, Feig C, Kultti A, Egeblad M, Fearon DT, Crawford JM, Clevers H, Park Y, Tuveson DA, Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer, J. Exp. Med 214 (2017) 579–596. 10.1084/jem.20162024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, Slagter M, van der Velden DL, Kaing S, Kelderman S, van Rooij N, van Leerdam ME, Depla A, Smit EF, Hartemink KJ, de Groot R, Wolkers MC, Sachs N, Snaebjornsson P, Monkhorst K, Haanen J, Clevers H, Schumacher TN, Voest EE, Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids, Cell. 174 (2018) 1586–1598.e12. 10.1016/j.cell.2018.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, Cantrell MA, Rack PG, Neal JT, Chan CWM, Yeung T, Gong X, Yuan J, Wilhelmy J, Robine S, Attardi LD, Plevritis SK, Hung KE, Chen CZ, Ji HP, Kuo CJ, Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture, Nat. Med 20 (2014) 769–777. 10.1038/nm.3585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Ootani A, Li X, Sangiorgi E, Ho QT, Ueno H, Toda S, Sugihara H, Fujimoto K, Weissman IL, Capecchi MR, Kuo CJ, Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche, Nat. Med 15 (2009) 701–706. 10.1038/nm.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, Deutsch BC, Liao L, Zemek AJ, Zhao F, Karlsson K, Schultz LM, Metzner TJ, Nadauld LD, Tseng YY, Alkhairy S, Oh C, Keskula P, Mendoza-Villanueva D, De La Vega FM, Kunz PL, Liao JC, Leppert JT, Sunwoo JB, Sabatti C, Boehm JS, Hahn WC, Zheng GXY, Davis MM, Kuo CJ, Organoid Modeling of the Tumor Immune Microenvironment, Cell. 175 (2018) 1972–1988.e16. 10.1016/j.cell.2018.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, Dowling C, Wanjala JN, Undvall EA, Arora VK, Wongvipat J, Kossai M, Ramazanoglu S, Barboza LP, Di W, Cao Z, Zhang QF, Sirota I, Ran L, Macdonald TY, Beltran H, Mosquera JM, Touijer KA, Scardino PT, Laudone VP, Curtis KR, Rathkopf DE, Morris MJ, Danila DC, Slovin SF, Solomon SB, Eastham JA, Chi P, Carver B, Rubin MA, Scher HI, Clevers H, Sawyers CL, Chen Y, Organoid cultures derived from patients with advanced prostate cancer, Cell. 159 (2014) 176–187. 10.1016/j.cell.2014.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Sato T, Stange DE, Ferrante M, Vries RGJ, Van Es JH, Van Den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, Clevers H, Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium, Gastroenterology. 141 (2011) 1762–1772. 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
  • [146].Weeber F, Ooft SN, Dijkstra KK, Voest EE, Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery, Cell Chem. Biol 24 (2017) 1092–1100. 10.1016/j.chembiol.2017.06.012. [DOI] [PubMed] [Google Scholar]
  • [147].Van De Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, Van Houdt W, Van Gorp J, Taylor-Weiner A, Kester L, McLaren-Douglas A, Blokker J, Jaksani S, Bartfeld S, Volckman R, Van Sluis P, Li VSW, Seepo S, Sekhar Pedamallu C, Cibulskis K, Carter SL, McKenna A, Lawrence MS, Lichtenstein L, Stewart C, Koster J, Versteeg R, Van Oudenaarden A, Saez-Rodriguez J, Vries RGJ, Getz G, Wessels L, Stratton MR, McDermott U, Meyerson M, Garnett MJ, Clevers H, Prospective derivation of a living organoid biobank of colorectal cancer patients, Cell. 161 (2015) 933–945. 10.1016/j.cell.2015.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Drost J, Van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, De Ligt J, Behjati S, Grolleman JE, Van Wezel T, Nik-Zainal S, Kuiper RP, Cuppen E, Clevers H, Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer, Science (80-. ). 358 (2017) 234–238. 10.1126/science.aao3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Rossi G, Manfrin A, Lutolf MP, Progress and potential in organoid research, Nat. Rev. Genet 19 (2018) 671–687. 10.1038/s41576-018-0051-9. [DOI] [PubMed] [Google Scholar]
  • [150].Yuki K, Cheng N, Nakano M, Kuo CJ, Organoid Models of Tumor Immunology, Trends Immunol. 41 (2020) 652–664. 10.1016/j.it.2020.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Fujii M, Sato T, Somatic cell-derived organoids as prototypes of human epithelial tissues and diseases, Nat. Mater 20 (2021) 156–169. 10.1038/s41563-020-0754-0. [DOI] [PubMed] [Google Scholar]
  • [152].Xu H, Lyu X, Yi M, Zhao W, Song Y, Wu K, Organoid technology and applications in cancer research, J. Hematol. Oncol (2018) 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC, Couce M, McLendon RE, Sloan AE, Rich JN, A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo, Cancer Res. 76 (2016) 2465–2477. 10.1158/0008-5472.CAN-15-2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Lee GY, Kenny PA, Lee EH, Bissell MJ, Three-dimensional culture models of normal and malignant breast epithelial cells, Nat. Methods 4 (2007) 359–365. 10.1038/nmeth1015.Three-dimensional. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Schoumacher M, Goldman RD, Louvard D, Vignjevic DM, Actin, microtubules, and vimentin intermediate filaments cooperate for elongation of invadopodia, J. Cell Biol 189 (2010) 541–556. 10.1083/jcb.200909113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Debnath J, Muthuswamy SK, Brugge JS, Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures, Methods. 30 (2003) 256–268. 10.1016/S1046-2023(03)00032-X. [DOI] [PubMed] [Google Scholar]
  • [157].Hotary K, Li XY, Allen E, Stevens SL, Weiss SJ, A cancer cell metalloprotease triad regulates the basement membrane transmigration program, Genes Dev. 20 (2006) 2673–2686. 10.1101/gad.1451806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Glentis A, Oertle P, Mariani P, Chikina A, El Marjou F, Attieh Y, Zaccarini F, Lae M, Loew D, Dingli F, Sirven P, Schoumacher M, Gurchenkov BG, Plodinec M, Vignjevic DM, Cancer-associated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane, Nat. Commun 8 (2017) 1–13. 10.1038/s41467-017-00985-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Cox TR, Erler JT, Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer, DMM Dis. Model. Mech 4 (2011) 165–178. 10.1242/dmm.004077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Micek HM, Visetsouk MR, Masters KS, Kreeger PK, Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment, IScience. 23 (2020) 101742. 10.1016/j.isci.2020.101742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Hall MS, Alisafaei F, Ban E, Feng X, Hui CY, Shenoy VB, Wu M, Fibrous nonlinear elasticity enables positive Mechanical feedback between cells and ECMs, Proc. Natl. Acad. Sci. U. S. A 113 (2016) 14043–14048. 10.1073/pnas.1613058113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Jordahl S, Solorio L, Neale DB, McDermott S, Jordahl JH, Fox A, Dunlay C, Xiao A, Brown M, Wicha M, Luker GD, Lahann J, Engineered Fibrillar Fibronectin Networks as Three- Dimensional Tissue Scaffolds, Adv. Mater 31 (2019) 1904580. 10.1002/adma.201904580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Burdick JA, Prestwich GD, Hyaluronic Acid Hydrogels for Biomedical Applications, Adv. Mater 23 (2011) 1–31. 10.1002/adma.201003963.Hyaluronic. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Wolf KJ, Lee S, Kumar S, A 3D topographical model of parenchymal infiltration and perivascular invasion in glioblastoma, APL Bioeng. 2 (2018). 10.1063/1.5021059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Pakshir P, Alizadehgiashi M, Wong B, Coelho NM, Chen X, Gong Z, Shenoy VB, McCulloch C, Hinz B, Dynamic fibroblast contractions attract remote macrophages in fibrillar collagen matrix, Nat. Commun 10 (2019) 1–17. 10.1038/s41467-019-09709-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Wu PH, Giri A, Sun SX, Wirtz D, Three-dimensional cell migration does not follow a random walk, Proc. Natl. Acad. Sci. U. S. A 111 (2014) 3949–3954. 10.1073/pnas.1318967111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Ilina O, Gritsenko PG, Syga S, Lippoldt J, La Porta CAM, Chepizhko O, Grosser S, Vullings M, Bakker GJ, Starruß J, Bult P, Zapperi S, Käs JA, Deutsch A, Friedl P, Cell–cell adhesion and 3D matrix confinement determine jamming transitions in breast cancer invasion, Nat. Cell Biol 22 (2020) 1103–1115. 10.1038/s41556-020-0552-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Seino T, Kawasaki S, Shimokawa M, Tamagawa H, Toshimitsu K, Fujii M, Ohta Y, Matano M, Nanki K, Kawasaki K, Takahashi S, Sugimoto S, Iwasaki E, Takagi J, Itoi T, Kitago M, Kitagawa Y, Kanai T, Sato T, Human Pancreatic Tumor Organoids Reveal Loss of Stem Cell Niche Factor Dependence during Disease Progression, Cell Stem Cell. 22 (2018) 454–467.e6. 10.1016/j.stem.2017.12.009. [DOI] [PubMed] [Google Scholar]
  • [169].Sapudom J, Rubner S, Martin S, Kurth T, Riedel S, Mierke CT, Pompe T, The phenotype of cancer cell invasion controlled by fibril diameter and pore size of 3D collagen networks, Biomaterials. 52 (2015) 367–375. 10.1016/j.biomaterials.2015.02.022. [DOI] [PubMed] [Google Scholar]
  • [170].Francis-sedlak ME, Uriel S, Larson JC, Greisler HP, Venerus DC, Brey EM, Characterization of type I collagen gels modified by glycation, Biomaterials. 30 (2009) 1851–1856. 10.1016/j.biomaterials.2008.12.014. [DOI] [PubMed] [Google Scholar]
  • [171].Roy R, Boskey A, Bonassar LJ, Processing of type I collagen gels using nonenzymatic glycation, J. Biomed. Mater. Res. - Part A 93 (2010) 843–851. 10.1002/jbm.a.32231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].NGUYEN-NGOC AJEK-V, Mammary ductal elongation and myoepithelial migration are regulated by the composition of the extracellular matrix K.-V., J. Microsc 251 (2013) 212–223. 10.1111/jmi.12017.Mammary. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Hofman K, Tucker N, Stanger J, Staiger M, Marshall S, Hall B, Effects of the molecular format of collagen on characteristics of electrospun fibres, J. Mater. Sci 47 (2012) 1148–1155. 10.1007/s10853-011-5775-2. [DOI] [Google Scholar]
  • [174].Guo C, K LJ. Ã, Flow and magnetic field induced collagen alignment, Biomaterials. 28 (2007) 1105–1114. 10.1016/j.biomaterials.2006.10.010. [DOI] [PubMed] [Google Scholar]
  • [175].Ray A, Slama ZM, Morford RK, Madden SA, Provenzano PP, Enhanced Directional Migration of Cancer Stem Cells in 3D Aligned Collagen Matrices, Biophys. J 112 (2017) 1023–1036. 10.1016/j.bpj.2017.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].McCoy MG, Wei JM, Choi S, Goerger JP, Zipfel W, Fischbach C, Collagen Fiber Orientation Regulates 3D Vascular Network Formation and Alignment, ACS Biomater. Sci. Eng 4 (2018) 2967–2976. 10.1021/acsbiomaterials.8b00384. [DOI] [PubMed] [Google Scholar]
  • [177].Lanfer B, Freudenberg U, Zimmermann R, Stamov D, Körber V, Werner C, Aligned fibrillar collagen matrices obtained by shear flow deposition, Biomaterials. 29 (2008) 3888–3895. 10.1016/j.biomaterials.2008.06.016. [DOI] [PubMed] [Google Scholar]
  • [178].Riching KM, Cox BL, Salick MR, Pehlke C, Riching AS, Ponik SM, Bass BR, Crone WC, Jiang Y, Weaver AM, Eliceiri KW, Keely PJ, 3D collagen alignment limits protrusions to enhance breast cancer cell persistence, Biophys. J 107 (2015) 2546–2558. 10.1016/j.bpj.2014.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Franco-Barraza J, Francescone R, Luong T, Shah N, Madhani R, Cukierman G, Dulaimi E, Devarajan K, Egleston BL, Nicolas E, Katherine Alpaugh R, Malik R, Uzzo RG, Hoffman JP, Golemis EA, Cukierman E, Matrix-regulated integrin αvβ5 maintains α5β1-dependent desmoplastic traits prognostic of neoplastic recurrence, Elife. 6 (2017) 1–46. 10.7554/eLife.20600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Amatangelo MD, Bassi DE, Klein-szanto JP, Cukierman E, Stroma-Derived Three-Dimensional Matrices Are Necessary and Sufficient to Promote Desmoplastic Differentiation of Normal Fibroblasts, Am. J. Pathol 167 (2005) 475–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Kaukonen R, Mai A, Georgiadou M, Saari M, De Franceschi N, Betz T, Sihto H, Elo L, Jokitalo E, Westermarck J, Normal stroma suppresses cancer cell proliferation via mechanosensitive regulation of JMJD1a-mediated transcription, Nat. Commun 7 (2016). 10.1038/ncomms12237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Franco-Barraza J, Raghavan KS, Luong T, Cukierman E, Engineering clinically-relevant human fibroblastic cell-derived extracellular matrices, in: Methods Cell Biol, 2020: pp. 109–160. 10.1016/bs.mcb.2019.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Seo BR, Bhardwaj P, Choi S, Gonzalez J, Eguiluz RCA, Wang K, Mohanan S, Morris PG, Du B, Zhou XK, Vahdat LT, Verma A, Elemento O, Hudis CA, Williams RM, Gourdon D, Dannenberg AJ, Fischbach C, Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis, Sci. Transl. Med 7 (2015). 10.1126/scitranslmed.3010467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Springer NL, Iyengar NM, Bareja R, Verma A, Jochelson MS, Giri DD, Zhou XK, Elemento O, Dannenberg AJ, Fischbach C, Obesity-Associated Extracellular Matrix Remodeling Promotes a Macrophage Phenotype Similar to Tumor-Associated Macrophages, Am. J. Pathol 189 (2019) 2019–2035. 10.1016/j.ajpath.2019.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Reichert JC, Quent VMC, Burke LJ, Stansfield SH, Clements JA, Hutmacher DW, Mineralized human primary osteoblast matrices as a model system to analyse interactions of prostate cancer cells with the bone microenvironment, Biomaterials. 31 (2010) 7928–7936. 10.1016/j.biomaterials.2010.06.055. [DOI] [PubMed] [Google Scholar]
  • [186].Choi S, Friedrichs J, Song YH, Werner C, Estroff LA, Fischbach C, Intrafibrillar, bone-mimetic collagen mineralization regulates breast cancer cell adhesion and migration, Biomaterials. 198 (2019) 95–106. 10.1016/j.biomaterials.2018.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Grayson WL, Fröhlich M, Yeager K, Bhumiratana S, Chan ME, Cannizzaro C, Engineering anatomically shaped human bone grafts, Proc. Natl. Acad. Sci 107 (2010) 1–6. 10.1073/pnas.0905439106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Hussey GS, Dziki JL, Badylak SF, Extracellular matrix-based materials for regenerative medicine, Nat. Rev. Mater 3 (2018) 159–173. 10.1038/s41578-018-0023-x. [DOI] [Google Scholar]
  • [189].Tian C, Öhlund D, Rickelt S, Lidström T, Huang Y, Hao L, Zhao RT, Franklin O, Bhatia SN, Tuveson DA, Hynes RO, Cancer cell–derived matrisome proteins promote metastasis in pancreatic ductal adenocarcinoma, Cancer Res. 80 (2020) 1461–1474. 10.1158/0008-5472.CAN-19-2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Tian X, Werner ME, Roche KC, Hanson AD, Foote HP, Yu SK, Warner SB, Copp JA, Lara H, Wauthier EL, Caster JM, Herring LE, Zhang L, Tepper JE, Hsu DS, Zhang T, Reid LM, Wang AZ, Organ-specific metastases obtained by culturing colorectal cancer cells on tissue-specific decellularized scaffolds, Nat. Biomed. Eng 2 (2018) 443–452. 10.1038/s41551-018-0231-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S, The use of whole organ decellularization for the generation of a vascularized liver organoid, Hepatology. 53 (2011) 604–617. 10.1002/hep.24067. [DOI] [PubMed] [Google Scholar]
  • [192].and W. Y Pelham Robert J., Cell locomotion and focal adhesions are regulated by substrate flexibility, Proc. Natl. Acad. Sci 94 (1997) 13661–13665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Tse JR, Engler AJ, Preparation of hydrogel substrates with tunable mechanical properties, Curr. Protoc. Cell Biol (2010). 10.1002/0471143030.cb1016s47. [DOI] [PubMed] [Google Scholar]
  • [194].Kapp TG, Rechenmacher F, Neubauer S, V Maltsev O, Cavalcanti-adam EA, Zarka R, Reuning U, Notni J, Wester H, Mas-moruno C, Spatz J, Geiger B, Kessler H, A Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins, Sci. Rep 7 (2017) 1–13. 10.1038/srep39805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Knight CG, Morton LF, Peachey AR, Tuckwell DS, Farndale RW, Barnes MJ, Chem MJJB, Recognize the Same Specific Amino Acid Sequence , GFOGER , in Native (Triple-helical) Collagens *, J. Biol. Chem 275 (2000) 35–40. 10.1074/jbc.275.1.35. [DOI] [PubMed] [Google Scholar]
  • [196].Raeber GP, Lutolf MP, Hubbell JA, Molecularly Engineered PEG Hydrogels : A Novel Model System for Proteolytically Mediated Cell Migration, Biophys. J 89 (2005) 1374–1388. 10.1529/biophysj.104.050682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Fong ELS, Wan X, Yang J, Morgado M, Mikos AG, Harrington DA, Navone NM, Farach-Carson MC, A 3D in vitro model of patient-derived prostate cancer xenograft for controlled interrogation of in vivo tumor-stromal interactions, Biomaterials. 77 (2016) 164–172. 10.1016/j.biomaterials.2015.10.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Vallmajo-Martin Q, Broguiere N, Millan C, Zenobi-Wong M, Ehrbar M, PEG/HA Hybrid Hydrogels for Biologically and Mechanically Tailorable Bone Marrow Organoids, Adv. Funct. Mater 30 (2020) 1–17. 10.1002/adfm.201910282. [DOI] [Google Scholar]
  • [199].Singh SP, Schwartz MP, Lee JY, Fairbanks BD, Anseth KS, A peptide functionalized poly(ethylene glycol) (PEG) hydrogel for investigating the influence of biochemical and biophysical matrix properties on tumor cell migration, Biomater. Sci 2 (2014) 1024–1034. 10.1039/c4bm00022f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Ehrbar M, Sala A, Lienemann P, Ranga A, Mosiewicz K, Bittermann A, Rizzi SC, Weber FE, Lutolf MP, Elucidating the role of matrix stiffness in 3D cell migration and remodeling, Biophys. J 100 (2011) 284–293. 10.1016/j.bpj.2010.11.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Nam S, Hu KH, Butte MJ, Chaudhuri O, Strain-enhanced stress relaxation impacts nonlinear elasticity in collagen gels, Proc. Natl. Acad. Sci. U. S. A 113 (2016) 5492–5497. 10.1073/pnas.1523906113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].McKinnon DD, Domaille DW, Cha JN, Anseth KS, Biophysically defined and cytocompatible covalently adaptable networks as viscoelastic 3d cell culture systems, Adv. Mater 26 (2014) 865–872. 10.1002/adma.201303680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Chaudhuri O, Gu L, Klumpers D, Darnell M, Bencherif SA, Weaver JC, Huebsch N, Lee HP, Lippens E, Duda GN, Mooney DJ, Hydrogels with tunable stress relaxation regulate stem cell fate and activity, Nat. Mater 15 (2016) 326–334. 10.1038/nmat4489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Bittner SM, Smith BT, Diaz-Gomez L, Hudgins CD, Melchiorri AJ, Scott DW, Fisher JP, Mikos AG, Fabrication and mechanical characterization of 3D printed vertical uniform and gradient scaffolds for bone and osteochondral tissue engineering, Acta Biomater. 90 (2019) 37–48. 10.1016/j.actbio.2019.03.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Angeloni V, Contessi N, De Marco C, Bertoldi S, Tanzi MC, Daidone MG, Farè S, Polyurethane foam scaffold as in vitro model for breast cancer bone metastasis, Acta Biomater. 63 (2017) 306–316. 10.1016/j.actbio.2017.09.017. [DOI] [PubMed] [Google Scholar]
  • [206].Martina Monique and Hutmacher Dietmar W, Biodegradable polymers applied in tissue engineering research: a review, Polym. Int 55 (2007) 145–157. 10.1002/pi. [DOI] [Google Scholar]
  • [207].Karageorgiou V, Kaplan D, Porosity of 3D biomaterial scaffolds and osteogenesis, Biomaterials. 26 (2005) 5474–5491. 10.1016/j.biomaterials.2005.02.002. [DOI] [PubMed] [Google Scholar]
  • [208].Fong ELS, Lamhamedi-Cherradi SE, Burdett E, Ramamoorthy V, Lazar AJ, Kasper FK, Farach-Carson MC, Vishwamitra D, Demicco EG, Menegaz BA, Amin HM, Mikos AG, Ludwig JA, Modeling Ewing sarcoma tumors in vitro with 3D scaffolds, Proc. Natl. Acad. Sci. U. S. A 110 (2013) 6500–6505. 10.1073/pnas.1221403110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, Polverini PJ, Mooney DJ, Engineering tumors with 3D scaffolds, Nat. Methods 4 (2007) 855–860. 10.1038/nmeth1085. [DOI] [PubMed] [Google Scholar]
  • [210].Murphy WL, Kohn DH, Mooney DJ, Growth of continuous bonelike mineral within porous poly(lactide-co- glycolide) scaffolds in vitro, J. Biomed. Mater. Res 50 (2000) 50–58. . [DOI] [PubMed] [Google Scholar]
  • [211].Boissard CIR, Bourban PE, Tami AE, Alini M, Eglin D, Nanohydroxyapatite/poly(ester urethane) scaffold for bone tissue engineering, Acta Biomater. 5 (2009) 3316–3327. 10.1016/j.actbio.2009.05.001. [DOI] [PubMed] [Google Scholar]
  • [212].Pathi SP, Lin DDW, Dorvee JR, Estroff LA, Fischbach C, Hydroxyapatite nanoparticle-containing scaffolds for the study of breast cancer bone metastasis, Biomaterials. 32 (2011) 5112–5122. 10.1016/j.biomaterials.2011.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Oakes RS, Bushnell GG, Orbach SM, Kandagatla P, Zhang Y, Morris AH, Hall MS, LaFaire P, Decker JT, Hartfield RM, Brooks MD, Wicha MS, Jeruss JS, Shea LD, Metastatic conditioning of myeloid cells at a subcutaneous synthetic niche reflects disease progression and predicts therapeutic outcomes, Cancer Res. 80 (2020) 602–612. 10.1158/0008-5472.CAN-19-1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Saha S, Duan X, Wu L, Lo P-K, Chen H, Wang Q, Electrospun Fibrous Scaffolds Promote Breast Cancer Cell Alignment and Epithelial–Mesenchymal Transition, Langmuir. 28 (2012) 2028–2034. 10.1021/la203846w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Baker BM, Trappmann B, Wang WY, Sakar MS, Kim IL, Shenoy VB, Burdick JA, Chen CS, Cell-mediated fibre recruitment drives extracellular matrix mechanosensing in engineered fibrillar microenvironments, Nat. Mater 14 (2015) 1262–1268. 10.1038/nmat4444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Azevedo H, Reis R, Understanding the Enzymatic Degradation of Biodegradable Polymers and Strategies to Control Their Degradation Rate, Biodegrad. Syst. Tissue Eng. Regen. Med (2004) 177–202. 10.1201/9780203491232.ch12. [DOI] [Google Scholar]
  • [217].Kim S, Kim HJ, Jeon NL, Biological applications of microfluidic gradient devices, Integr. Biol 2 (2010) 584. 10.1039/c0ib00055h. [DOI] [PubMed] [Google Scholar]
  • [218].Dertinger SKW, Chiu DT, Jeon NL, Whitesides GM, Generation of Gradients Having Complex Shapes Using Microfluidic Networks, Anal. Chem 73 (2001) 1240–1246. 10.1021/ac001132d. [DOI] [Google Scholar]
  • [219].Cavnar SP, Ray P, Moudgil P, Chang SL, Luker KE, Linderman JJ, Takayama S, Luker GD, Microfluidic source-sink model reveals effects of biophysically distinct CXCL12 isoforms in breast cancer chemotaxis, Integr. Biol 6 (2014) 564–576. 10.1039/C4IB00015C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Mi S, Du Z, Xu Y, Wu Z, Qian X, Zhang M, Sun W, Microfluidic co-culture system for cancer migratory analysis and anti-metastatic drugs screening, Sci. Rep 6 (2016) 1–11. 10.1038/srep35544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Berthier E, Beebe DJ, Gradient generation platforms: new directions for an established microfluidic technology, Lab Chip. 14 (2014) 3241–3247. 10.1039/C4LC00448E. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Zervantonakis I, Chung SSS, Sudo R, Zhang M, Charest J, Kamm R, Barbara S, Concentration gradients in microfluidic 3D matrix cell culture systems, Int. J. Micro-Nano Scale Transp 1 (2010) 27–36. 10.1260/1759-3093.1.1.27. [DOI] [Google Scholar]
  • [223].Hwang PY, Brenot A, King AC, Longmore GD, George SC, Randomly Distributed K14 + Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues, Cancer Res. 79 (2019) 1899–1912. 10.1158/0008-5472.CAN-18-2828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Ayuso JM, Gillette A, Lugo-Cintrón K, Acevedo-Acevedo S, Gomez I, Morgan M, Heaster T, Wisinski KB, Palecek SP, Skala MC, Beebe DJ, Organotypic microfluidic breast cancer model reveals starvation-induced spatial-temporal metabolic adaptations, EBioMedicine. 37 (2018) 144–157. 10.1016/j.ebiom.2018.10.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Ayuso JM, Virumbrales-munoz M, McMinn PH, Rehman S, Gomez I, Karim MR, Trusttchel R, Wisinski KB, Beebe DJ, Skala MC, Karim DMR, Trusttchel R, Wisinski KB, Beebe J, Skala MC, Karim MR, Trusttchel R, Wisinski KB, Beebe DJ, Skala MC, Tumor-on-a-chip: a microfluidic model to study cell response to environmental gradients, Lab Chip. 19 (2019) 3461–3471. 10.1039/c9lc00270g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Lewis DM, Park KM, Tang V, Xu Y, Pak K, Eisinger-Mathason TSK, Simon MC, Gerecht S, Intratumoral oxygen gradients mediate sarcoma cell invasion, Proc. Natl. Acad. Sci 113 (2016) 9292–9297. 10.1073/pnas.1605317113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Lewis DM, Blatchley MR, Park KM, Gerecht S, O2-controllable hydrogels for studying cellular responses to hypoxic gradients in three dimensions in vitro and in vivo, Nat. Protoc 12 (2017) 1620–1638. 10.1038/nprot.2017.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Rodenhizer D, Gaude E, Cojocari D, Mahadevan R, Frezza C, Wouters BG, McGuigan AP, A three-dimensional engineered tumour for spatial snapshot analysis of cell metabolism and phenotype in hypoxic gradients, Nat. Mater 15 (2016) 227–234. 10.1038/nmat4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Young M, Rodenhizer D, Dean T, D’Arcangelo E, Xu B, Ailles L, McGuigan AP, A TRACER 3D Co-Culture tumour model for head and neck cancer, Biomaterials. 164 (2018) 54–69. 10.1016/j.biomaterials.2018.01.038. [DOI] [PubMed] [Google Scholar]
  • [230].Dean T, Li NT, Cadavid JL, Ailles L, McGuigan AP, A TRACER culture invasion assay to probe the impact of cancer associated fibroblasts on head and neck squamous cell carcinoma cell invasiveness, Biomater. Sci 8 (2020) 3078–3094. 10.1039/c9bm02017a. [DOI] [PubMed] [Google Scholar]
  • [231].Bonvin C, Overney J, Shieh AC, Dixon JB, Swartz MA, A multichamber fluidic device for 3D cultures under interstitial flow with live imaging: Development, characterization, and applications, Biotechnol. Bioeng 105 (2010) 982–991. 10.1002/bit.22608. [DOI] [PubMed] [Google Scholar]
  • [232].Haessler U, Teo JCM, Foretay D, Renaud P, Swartz MA, Migration dynamics of breast cancer cells in a tunable 3D interstitial flow chamber, Integr. Biol 4 (2012) 401–409. 10.1039/c1ib00128k. [DOI] [PubMed] [Google Scholar]
  • [233].Polacheck WJ, German AE, Mammoto A, Ingber DE, Kamm RD, Mechanotransduction of fluid stresses governs 3D cell migration, Proc. Natl. Acad. Sci 111 (2014) 2447–2452. 10.1073/pnas.1316848111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Alonzo LF, Moya ML, Shirure VS, George SC, Microfluidic device to control interstitial flow-mediated homotypic and heterotypic cellular communication, Lab Chip. 15 (2015) 3521–3529. 10.1039/C5LC00507H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Munson JM, Bellamkonda RV, Swartz MA, Interstitial Flow in a 3D Microenvironment Increases Glioma Invasion by a CXCR4-Dependent Mechanism, Cancer Res. 73 (2013) 1536–1546. 10.1158/0008-5472.CAN-12-2838. [DOI] [PubMed] [Google Scholar]
  • [236].Cornelison RC, Brennan CE, Kingsmore KM, Munson JM, Convective forces increase CXCR4-dependent glioblastoma cell invasion in GL261 murine model, Sci. Rep 8 (2018) 1–11. 10.1038/s41598-018-35141-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Shields JD, Fleury ME, Yong C, Tomei AA, Randolph GJ, Swartz MA, Autologous Chemotaxis as a Mechanism of Tumor Cell Homing to Lymphatics via Interstitial Flow and Autocrine CCR7 Signaling, Cancer Cell. 11 (2007) 526–538. 10.1016/j.ccr.2007.04.020. [DOI] [PubMed] [Google Scholar]
  • [238].Tien J, Dance YW, Ghani U, Seibel AJ, Nelson CM, Interstitial Hypertension Suppresses Escape of Human Breast Tumor Cells Via Convection of Interstitial Fluid, Cell. Mol. Bioeng (2020). 10.1007/s12195-020-00661-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Lewis DM, Gerecht S, Microfluidics and biomaterials to study angiogenesis, Curr. Opin. Chem. Eng 11 (2016) 114–122. 10.1016/j.coche.2016.02.005. [DOI] [Google Scholar]
  • [240].Farahat WA, Wood LB, Zervantonakis IK, Schor A, Ong S, Neal D, Kamm RD, Asada HH, Ensemble analysis of angiogenic growth in three-dimensional microfluidic cell cultures, PLoS One. 7 (2012) e37333. 10.1371/journal.pone.0037333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Chen MB, Whisler JA, Fröse J, Yu C, Shin Y, Kamm RD, On-chip human microvasculature assay for visualization and quantification of tumor cell extravasation dynamics, Nat. Protoc 12 (2017) 865–880. 10.1038/nprot.2017.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Song JW, Bazou D, Munn LL, Anastomosis of endothelial sprouts forms new vessels in a tissue analogue of angiogenesis, Integr. Biol 4 (2012) 857. 10.1039/c2ib20061a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Song JW, Munn LL, Fluid forces control endothelial sprouting, Proc. Natl. Acad. Sci 108 (2011) 15342–15347. 10.1073/pnas.1105316108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Zervantonakis IK, Hughes-Alford SK, Charest JL, Condeelis JS, Gertler FB, Kamm RD, Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function, Proc. Natl. Acad. Sci 109 (2012) 13515–13520. 10.1073/pnas.1210182109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Brett M-E, Bomberger HE, Doak GR, Price MA, McCarthy JB, Wood DK, In vitro elucidation of the role of pericellular matrix in metastatic extravasation and invasion of breast carcinoma cells, Integr. Biol 10 (2018) 242–252. 10.1039/C7IB00173H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino M, Fessler E, Chen MB, Krall JA, DeCock J, Zervantonakis IK, Iannello A, Iwamoto Y, Cortez-Retamozo V, Kamm RD, Pittet MJ, Raulet DH, Weinberg RA, Neutrophils Suppress Intraluminal NK Cell–Mediated Tumor Cell Clearance and Enhance Extravasation of Disseminated Carcinoma Cells, Cancer Discov. 6 (2016) 630–649. 10.1158/2159-8290.CD-15-1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Truong D, Puleo J, Llave A, Mouneimne G, Kamm RD, Nikkhah M, Breast Cancer Cell Invasion into a Three Dimensional Tumor-Stroma Microenvironment, Sci. Rep 6 (2016) 34094. 10.1038/srep34094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Nagaraju S, Truong D, Mouneimne G, Nikkhah M, Microfluidic Tumor-Vascular Model to Study Breast Cancer Cell Invasion and Intravasation, Adv. Healthc. Mater 7 (2018) 1701257. 10.1002/adhm.201701257. [DOI] [PubMed] [Google Scholar]
  • [249].Chrobak KM, Potter DR, Tien J, Formation of perfused, functional microvascular tubes in vitro, Microvasc. Res 71 (2006) 185–196. 10.1016/j.mvr.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • [250].Polacheck WJ, Kutys ML, Tefft JB, Chen CS, Microfabricated blood vessels for modeling the vascular transport barrier, Springer; US, 2019. 10.1038/s41596-019-0144-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Wiedeman MP, Dimensions of Blood Vessels from Distributing Artery to Collecting Vein, Circ. Res 12 (1963) 375–378. 10.1161/01.RES.12.4.375. [DOI] [PubMed] [Google Scholar]
  • [252].Morgan JP, Delnero PF, Zheng Y, Verbridge SS, Chen J, Craven M, Choi NW, Diaz-Santana A, Kermani P, Hempstead B, López JA, Corso TN, Fischbach C, Stroock AD, Formation of microvascular networks in vitro, Nat. Protoc 8 (2013) 1820–1836. 10.1038/nprot.2013.110. [DOI] [PubMed] [Google Scholar]
  • [253].Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT, Cohen DM, Toro E, Chen AA, Galie PA, Yu X, Chaturvedi R, Bhatia SN, Chen CS, Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues, Nat. Mater 11 (2012) 768–774. 10.1038/nmat3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Kotha SS, Hayes BJ, Phong KT, Redd MA, Bomsztyk K, Ramakrishnan A, Torok-Storb B, Zheng Y, Engineering a multicellular vascular niche to model hematopoietic cell trafficking, Stem Cell Res. Ther 9 (2018) 77. 10.1186/s13287-018-0808-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Zheng Y, Chen J, Craven M, Choi NW, Totorica S, Diaz-Santana A, Kermani P, Hempstead B, Fischbach-Teschl C, Lopez JA, Stroock AD, In vitro microvessels for the study of angiogenesis and thrombosis, Proc. Natl. Acad. Sci 109 (2012) 9342–9347. 10.1073/pnas.1201240109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Wang WY, Lin D, Jarman EH, Polacheck WJ, Baker BM, Functional angiogenesis requires microenvironmental cues balancing endothelial cell migration and proliferation, Lab Chip. 20 (2020) 1153–1166. 10.1039/c9lc01170f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Nguyen D-HT, Lee E, Alimperti S, Norgard RJ, Wong A, Lee JJ-K, Eyckmans J, Stanger BZ, Chen CS, A biomimetic pancreatic cancer on-chip reveals endothelial ablation via ALK7 signaling, Sci. Adv 5 (2019) eaav6789. 10.1126/sciadv.aav6789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Sobrino A, Phan DTT, Datta R, Wang X, Hachey SJ, Romero-López M, Gratton E, Lee AP, George SC, Hughes CCW, 3D microtumors in vitro supported by perfused vascular networks, Sci. Rep 6 (2016) 31589. 10.1038/srep31589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Chen MB, Lamar JM, Li R, Hynes RO, Kamm RD, Elucidation of the roles of tumor integrin β1 in the extravasation stage of the metastasis cascade, Cancer Res. 76 (2016) 2513–2524. 10.1158/0008-5472.CAN-15-1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Palikuqi B, Nguyen D-HT, Li G, Schreiner R, Pellegata AF, Liu Y, Redmond D, Geng F, Lin Y, Gómez-Salinero JM, Yokoyama M, Zumbo P, Zhang T, Kunar B, Witherspoon M, Han T, Tedeschi AM, Scottoni F, Lipkin SM, Dow L, Elemento O, Xiang JZ, Shido K, Spence JR, Zhou QJ, Schwartz RE, De Coppi P, Rabbany SY, Rafii S, Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis, Nature. 585 (2020) 426–432. 10.1038/s41586-020-2712-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Bhatia SN, Ingber DE, Microfluidic organs-on-chips, Nat. Biotechnol 32 (2014) 760–772. 10.1038/nbt.2989. [DOI] [PubMed] [Google Scholar]
  • [262].Park TE, Mustafaoglu N, Herland A, Hasselkus R, Mannix R, FitzGerald EA, Prantil-Baun R, Watters A, Henry O, Benz M, Sanchez H, McCrea HJ, Goumnerova LC, Song HW, Palecek SP, Shusta E, Ingber DE, Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies, Nat. Commun 10 (2019) 1–12. 10.1038/s41467-019-10588-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Chen WLK, Edington C, Suter E, Yu J, Velazquez JJ, Velazquez JG, Shockley M, Large EM, Venkataramanan R, Hughes DJ, Stokes CL, Trumper DL, Carrier RL, Cirit M, Griffith LG, Lauffenburger DA, Integrated gut/liver microphysiological systems elucidates inflammatory inter-tissue crosstalk, Biotechnol. Bioeng 114 (2017) 2648–2659. 10.1002/bit.26370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Jalili-Firoozinezhad S, Gazzaniga FS, Calamari EL, Camacho DM, Fadel CW, Bein A, Swenor B, Nestor B, Cronce MJ, Tovaglieri A, Levy O, Gregory KE, Breault DT, Cabral JMS, Kasper DL, Novak R, Ingber DE, A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip, Nat. Biomed. Eng 3 (2019) 520–531. 10.1038/s41551-019-0397-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Choi Y, Hyun E, Seo J, Blundell C, Kim HC, Lee E, Lee SH, Moon A, Moon WK, Huh D, A microengineered pathophysiological model of early-stage breast cancer, Lab Chip. 15 (2015) 3350–3357. 10.1039/C5LC00514K. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Bi Y, Shirure VS, Liu R, Cunningham C, Ding L, Meacham JM, Goedegebuure SP, George SC, Fields RC, Tumor-on-a-chip platform to interrogate the role of macrophages in tumor progression, Integr. Biol 12 (2020) 221–232. 10.1093/intbio/zyaa017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Clark AM, Wheeler SE, Young CL, Stockdale L, Shepard Neiman J, Zhao W, Stolz DB, Venkataramanan R, Lauffenburger D, Griffith L, Wells A, A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties, Lab Chip. 17 (2017) 156–168. 10.1039/C6LC01171C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Torisawa Y, Spina CS, Mammoto T, Mammoto A, Weaver JC, Tat T, Collins JJ, Ingber DE, Bone marrow–on–a–chip replicates hematopoietic niche physiology in vitro, Nat. Methods 11 (2014) 663–669. 10.1038/nmeth.2938. [DOI] [PubMed] [Google Scholar]
  • [269].Marturano-Kruik A, Nava MM, Yeager K, Chramiec A, Hao L, Robinson S, Guo E, Raimondi MT, Vunjak-Novakovic G, Human bone perivascular niche-on-a-chip for studying metastatic colonization, Proc. Natl. Acad. Sci 115 (2018) 1256–1261. 10.1073/pnas.1714282115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE, Reconstituting Organ-Level Lung Functions on a Chip, Science (80-. ). 328 (2010) 1662–1668. 10.1126/science.1188302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Hassell BA, Goyal G, Lee E, Sontheimer-Phelps A, Levy O, Chen CS, Ingber DE, Human Organ Chip Models Recapitulate Orthotopic Lung Cancer Growth, Therapeutic Responses, and Tumor Dormancy In Vitro, Cell Rep. 21 (2017) 508–516. 10.1016/j.celrep.2017.09.043. [DOI] [PubMed] [Google Scholar]
  • [272].Sontheimer-Phelps A, Hassell BA, Ingber DE, Phelps AS, Modelling cancer in microfluidic human organs-on-chips, Nat. Rev. Cancer 19 (2019) 65–81. 10.1038/s41568-018-0104-6. [DOI] [PubMed] [Google Scholar]
  • [273].Chramiec A, Teles D, Yeager K, Marturano-Kruik A, Pak J, Chen T, Hao L, Wang M, Lock R, Tavakol DN, Lee MB, Kim J, Ronaldson-Bouchard K, Vunjak-Novakovic G, Integrated human organ-on-a-chip model for predictive studies of anti-tumor drug efficacy and cardiac safety, Lab Chip. 20 (2020) 4357–4372. 10.1039/D0LC00424C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Weng K-C, Kurokawa YK, Hajek BS, Paladin JA, Shirure VS, George SC, Human Induced Pluripotent Stem-Cardiac-Endothelial-Tumor-on-a-Chip to Assess Anticancer Efficacy and Cardiotoxicity, Tissue Eng. Part C Methods 26 (2020) 44–55. 10.1089/ten.tec.2019.0248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Schepers A, Li C, Chhabra A, Seney BT, Bhatia S, Engineering a perfusable 3D human liver platform from iPS cells, Lab Chip. 16 (2016) 2644–2653. 10.1039/C6LC00598E. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Novak R, Ingram M, Marquez S, Das D, Delahanty A, Herland A, Maoz BM, Jeanty SSF, Somayaji MR, Burt M, Calamari E, Chalkiadaki A, Cho A, Choe Y, Chou DB, Cronce M, Dauth S, Divic T, Fernandez-Alcon J, Ferrante T, Ferrier J, FitzGerald EA, Fleming R, Jalili-Firoozinezhad S, Grevesse T, Goss JA, Hamkins-Indik T, Henry O, Hinojosa C, Huffstater T, Jang KJ, Kujala V, Leng L, Mannix R, Milton Y, Nawroth J, Nestor BA, Ng CF, O’Connor B, Park TE, Sanchez H, Sliz J, Sontheimer-Phelps A, Swenor B, Thompson G, Touloumes GJ, Tranchemontagne Z, Wen N, Yadid M, Bahinski A, Hamilton GA, Levner D, Levy O, Przekwas A, Prantil-Baun R, Parker KK, Ingber DE, Robotic fluidic coupling and interrogation of multiple vascularized organ chips, Nat. Biomed. Eng 4 (2020) 407–420. 10.1038/s41551-019-0497-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Edington CD, Chen WLK, Geishecker E, Kassis T, Soenksen LR, Bhushan BM, Freake D, Kirschner J, Maass C, Tsamandouras N, Valdez J, Cook CD, Parent T, Snyder S, Yu J, Suter E, Shockley M, Velazquez J, Velazquez JJ, Stockdale L, Papps JP, Lee I, Vann N, Gamboa M, LaBarge ME, Zhong Z, Wang X, Boyer LA, Lauffenburger DA, Carrier RL, Communal C, Tannenbaum SR, Stokes CL, Hughes DJ, Rohatgi G, Trumper DL, Cirit M, Griffith LG, Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies, Sci. Rep 8 (2018) 4530. 10.1038/s41598-018-22749-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [278].Shirure VS, Bi Y, Curtis MB, Lezia A, Goedegebuure MM, Goedegebuure SP, Aft R, Fields RC, George SC, Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids, Lab Chip. 18 (2018) 3687–3702. 10.1039/C8LC00596F. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Carter S-SD, Atif A-R, Kadekar S, Lanekoff I, Engqvist H, Varghese OP, Tenje M, Mestres G, PDMS leaching and its implications for on-chip studies focusing on bone regeneration applications, Organs-on-a-Chip. 2 (2020) 100004. 10.1016/j.ooc.2020.100004. [DOI] [Google Scholar]
  • [280].Albritton JL, Miller JS, 3D bioprinting: Improving in vitro models of metastasis with heterogeneous tumor microenvironments, DMM Dis. Model. Mech 10 (2017) 3–14. 10.1242/dmm.025049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Jeon O, Lee YB, Hinton TJ, Feinberg AW, Alsberg E, Cryopreserved cell-laden alginate microgel bioink for 3D bioprinting of living tissues, Mater. Today Chem 12 (2019) 61–70. 10.1016/j.mtchem.2018.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Lee A, Hudson AR, Shiwarski DJ, Tashman JW, Hinton TJ, Yerneni S, Bliley JM, Campbell PG, Feinberg AW, 3D bioprinting of collagen to rebuild components of the human heart, Science (80-. ). 365 (2019) 482–487. 10.1126/science.aav9051. [DOI] [PubMed] [Google Scholar]
  • [283].Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Shue H-J, Ramadan MH, Hudson AR, Feinberg AW, Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels, Sci. Adv 1 (2015) e1500758. 10.1126/sciadv.1500758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Daly AC, Davidson MD, Burdick JA, 3D bioprinting of high cell-density heterogeneous tissue models through spheroid fusion within self-healing hydrogels, Nat. Commun 12 (2021) 1–13. 10.1038/s41467-021-21029-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Hinton TJ, Lee A, Feinberg AW, 3D bioprinting from the micrometer to millimeter length scales: Size does matter, Curr. Opin. Biomed. Eng 1 (2017) 31–37. 10.1016/j.cobme.2017.02.004. [DOI] [Google Scholar]
  • [286].Grigoryan B, Paulsen SJ, Corbett DC, Sazer DW, Fortin CL, Zaita AJ, Greenfield PT, Calafat NJ, Gounley JP, Ta AH, Johansson F, Randles A, Rosenkrantz JE, Louis-Rosenberg JD, Galie PA, Stevens KR, Miller JS, Multivascular networks and functional intravascular topologies within biocompatible hydrogels, Science (80-. ). 364 (2019) 458–464. 10.1126/science.aav9750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Kinstlinger IS, Saxton SH, Calderon GA, Ruiz KV, Yalacki DR, Deme PR, Rosenkrantz JE, Louis-Rosenberg JD, Johansson F, Janson KD, Sazer DW, Panchavati SS, Bissig K-D, Stevens KR, Miller JS, Generation of model tissues with dendritic vascular networks via sacrificial laser-sintered carbohydrate templates, Nat. Biomed. Eng 4 (2020) 916–932. 10.1038/s41551-020-0566-1. [DOI] [PubMed] [Google Scholar]
  • [288].Skylar-Scott MA, Uzel SGM, Nam LL, Ahrens JH, Truby RL, Damaraju S, Lewis JA, Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels, Sci. Adv 5 (2019). 10.1126/sciadv.aaw2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [289].Homan KA, Kolesky DB, Skylar-Scott MA, Herrmann J, Obuobi H, Moisan A, Lewis JA, Bioprinting of 3D Convoluted Renal Proximal Tubules on Perfusable Chips, Sci. Rep 6 (2016) 34845. 10.1038/srep34845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [290].Paulsen SJ, Miller JS, Tissue vascularization through 3D printing: Will technology bring us flow?, Dev. Dyn 244 (2015) 629–640. 10.1002/dvdy.24254. [DOI] [PubMed] [Google Scholar]
  • [291].Pati F, Jang J, Ha D-H, Won Kim S, Rhie J-W, Shim J-H, Kim D-H, Cho D-W, Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink, Nat. Commun 5 (2014) 3935. 10.1038/ncomms4935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [292].Grolman JM, Zhang D, Smith AM, Moore JS, Kilian KA, Rapid 3D Extrusion of Synthetic Tumor Microenvironments, Adv. Mater 27 (2015) 5512–5517. 10.1002/adma.201501729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [293].Camacho P, Busari H, Seims KB, Schwarzenberg P, Dailey HL, Chow LW, 3D printing with peptide–polymer conjugates for single-step fabrication of spatially functionalized scaffolds, Biomater. Sci 7 (2019) 4237–4247. 10.1039/C9BM00887J. [DOI] [PubMed] [Google Scholar]
  • [294].Kaushik G, Gil DA, Torr E, Berge ES, Soref C, Uhl P, Fontana G, Antosiewicz-Bourget J, Edington C, Schwartz MP, Griffith LG, Thomson JA, Skala MC, Daly WT, Murphy WL, Quantitative Label-Free Imaging of 3D Vascular Networks Self-Assembled in Synthetic Hydrogels, Adv. Healthc. Mater 8 (2019) 1801186. 10.1002/adhm.201801186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Gil DA, Deming D, Skala MC, Patient-derived cancer organoid tracking with wide-field one-photon redox imaging to assess treatment response, J. Biomed. Opt 26 (2021) 1–19. 10.1117/1.JBO.26.3.036005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Mulligan JA, Ling L, Leartprapun N, Fischbach C, Adie SG, Computational 4D-OCM for label-free imaging of collective cell invasion and force-mediated deformations in collagen, Sci. Rep 11 (2021) 2814. 10.1038/s41598-021-81470-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Liu Y, Yang M, Deng Y, Su G, Enninful A, Guo CC, Tebaldi T, Zhang D, Kim D, Bai Z, Norris E, Pan A, Li J, Xiao Y, Halene S, Fan R, High-Spatial-Resolution Multi-Omics Sequencing via Deterministic Barcoding in Tissue, Cell. 183 (2020) 1665–1681.e18. 10.1016/j.cell.2020.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Liu Y, Yang M, Deng Y, Su G, Enninful A, Guo CC, Tebaldi T, Zhang D, Kim D, Bai Z, Norris E, Pan A, Li J, Xiao Y, Halene S, Fan R, High-Spatial-Resolution Multi-Omics Sequencing via Deterministic Barcoding in Tissue, Cell. 183 (2020) 1665–1681.e18. 10.1016/j.cell.2020.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Robertson-Tessi M, Gillies RJ, Gatenby RA, Anderson ARA, Impact of Metabolic Heterogeneity on Tumor Growth, Invasion, and Treatment Outcomes, Cancer Res. 75 (2015) 1567–1579. 10.1158/0008-5472.CAN-14-1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Rejniak KA, Anderson ARA, Hybrid models of tumor growth, Wiley Interdiscip. Rev. Syst. Biol. Med 3 (2011) 115–125. 10.1002/wsbm.102. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES