Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 4.
Published in final edited form as: Ann Biomed Eng. 2019 Oct 15;48(7):2064–2077. doi: 10.1007/s10439-019-02384-0

Emerging Biomimetic Materials for Studying Tumor and Immune Cell Behavior

LOGAN A NORTHCUTT 1, ALEJANDRA SUAREZ-ARNEDO 2, MARJAN RAFAT 1,3,4,5
PMCID: PMC7156320  NIHMSID: NIHMS1057109  PMID: 31617045

Abstract

Cancer is one of the leading causes of death both in the United States and worldwide. The dynamic microenvironment in which tumors grow consists of fibroblasts, immune cells, extracellular matrix (ECM), and cytokines that enable progression and metastasis. Novel biomaterials that mimic these complex surroundings give insight into the biological, chemical, and physical environment that cause cancer cells to metastasize and invade in to other tissues. Two-dimensional (2D) cultures are useful for gaining limited information about cancer cell behavior; however, they do not accurately represent the environments that cells experience in vivo. Recent advances in the design and tunability of diverse three-dimensional (3D) biomaterials complement biological knowledge and allow for improved recapitulation of in vivo conditions. Understanding cell–ECM and cell–cell interactions that facilitate tumor survival will accelerate the design of more effective therapies. This review discusses innovative materials currently being used to study tumor and immune cell behavior and interactions, including materials that mimic the ECM composition, mechanical stiffness, and integrin binding sites of the tumor microenvironment.

Keywords: Biomimetic, Extracellular matrix, Epithelial-to-mesenchymal transition, Cell–ECM interactions

INTRODUCTION

The ability of tumor cells to metastasize beyond the primary tumor and invade into new tissues is one highly-studied hallmark of cancer.44 To gain more information about this phenomenon, physicians and researchers use techniques such as incisional biopsies to extract tumor samples. These specimens are helpful for gaining information such as expression of genes and prevalent proteins that are associated with malignant phenotypes and cancer cell proliferation.99 Although informative and extracted from patients, one disadvantage of direct evaluation of primary tissues is the large degree of variability in each sample. Another popular methodology in studying cancer in vitro is two-dimensional (2D) tissue culture. One key limitation of this method is that 2D culture does not recapitulate physiologically relevant three-dimensional (3D) microenvironments, which typically results in distinct outcomes from in vivo conditions.80 Another drawback of this technique is primarily the lack of integrin binding sites present in the tumor microenvironment. In addition, the complexity and tunability of 2D environments are severely limited.14

While much information has been gained from monolayer systems, there has been a recent push to develop tunable, 3D cell-culturing systems to better mimic the tumor microenvironment, which includes fibroblasts, immune cells, soluble factors, and extracellular matrix (ECM) proteins (Fig. 1). One of the most sought-after approaches to address these issues is the use of biomimetic materials that model specific in vivo characteristics, including chemical composition or stiffness.35 Biomimetic materials have been developed for applications ranging from the regeneration of cartilage to wound healing.2 Currently, these materials have also been used to create synthetic environments that can be used to study cell morphology as well as the underlying molecular causes of malignant cell morphology and invasion.

FIGURE 1.

FIGURE 1.

The tumor microenvironment and its components, including tumor cells, immune cells, stromal cells, ECM, and soluble factors.

In addition to tumor cells alone, incorporating multiple cell types such as immune and stromal cell populations have allowed researchers to better model the in vivo microenvironment, cell–cell interactions, and cell–ECM interactions. This has led to an increased understanding of tumor cell behavior and improved tumor targeting therapeutic strategies. The immunological response is also an important factor when developing 3D microenvironments. Tumor-promoting inflammation is an additional hallmark of cancer, so understanding how immune cells interact with tumor cells in the microenvironment is imperative for determining the causes behind tumor progression and metastasis.36,44,123 In this review, we will outline the emerging materials utilized in designing biomimetic microenvironments for evaluating tumor and immune cell behavior.

THE EXTRACELLULAR MATRIX (ECM)

Defining the ECM

The ECM is a 3D network that consists of various proteins and macromolecules necessary for providing cellular, biochemical, and structural support. All tissues in the body contain ECM components but vary in their composition. Most mammalian ECM consists of collagen, fibronectin, and laminin, along with glycosaminoglycans that form proteoglycans.33,124 Table 1 lists the key components present in the ECM. In cancer studies, cell–ECM interactions are commonly studied (Fig. 2), which facilitate changes in cytoskeletal actin filaments, upregulation or downregulation of oncogenes, and expression of proteins associated with cancer cell invasion and proliferation.11,14

TABLE 1.

Components of the ECM.

Main components Macromolecules Function
Highly viscous proteoglycans144 Aggrecan Interact with integrins
Decorin Hydration buffer
Lumican Mitigate shear stress
Perlecan
Syndecan
Versican
Insoluble collagen fibers30,144 Collagen I Provide structure
Collagen II Cell adhesion
Collagen III
Collagen IV
Collagen V
Soluble multiadhesive proteins1,43,144 Laminin Integrin binding
Fibronectin Cell growth
Cell adhesion

FIGURE 2.

FIGURE 2.

ECM-cell interactions.

ECM and the Tumor Microenvironment

The ECM is a prominent factor in cancer progression in the tumor microenvironment.7 There has been extensive research into how the chemical and physical properties of the ECM can affect the proliferation and migration of cancer and immune cells as the composition and structure of the ECM is integral to the regulation of cell growth, phenotype, and organization.131 Many recent studies have attempted to mimic particular ECM properties and structures to study cancer progression. For example, Narkhede et al. developed hyaluronic acid (HA)-based hydrogels since HA is a key component of the brain ECM and has similar viscoelastic properties to the ECM.94 The HA hydrogel stiffness was varied from 0.2 to 4.5 kPa to understand how changes in mechanical cues can affect the behavior of brain metastatic breast cancer cells. According to their results, breast cancer cells proliferate and adhere to a greater extent in stiffer microenvironments, and this was mediated by the focal adhesion kinase (FAK)–phosphoinositide-3 kinase (PI3K) pathway.94 Another example is the work by Kim et al. where they incorporated collagen I and Matrigel into a microfluidic device to study how the recruitment of immune cells alters the invasiveness of cancer cells. This work demonstrated monocyte and macrophage-induced remodeling of the ECM, which enhanced the invasiveness of cancer cells.63 ECM components have also been utilized as bioinks in 3D printing technologies.46,71 For instance, Duarte Campos et al. used a type I collagen-based bioink to make a 3D mini-model of neuroblastoma. These models were able to mimic in vivo characteristics of neuroblastoma such as proliferative potential and the formation of Homer Wright-like rosettes.27,134

Integrin Binding Patterns

One of the most important drivers of communication between cells and the ECM is the presence of integrins. Integrins have key functions in multiple processes that facilitate tumorigenesis, progression, and metastatic potential of cancer cells.58 These cellular receptors are implicated in signaling molecules, cell migration machinery, the epithelial-to-mesenchymal transition (EMT), and mechanotransduction.42,133 For instance, integrins allow for the activation of epidermal growth factor receptor (EGFR) and the modulation of epithelial differentiation in 3D microenvironments. EGFR also promotes the formation of focal adhesions, structures that bind integrins to the cytoskeleton and organize the conformation, tension, and morphology of cells.52 Integrin αvβ6 is the binding receptor in tumorigenic cells while α5β1 is a constitutively expressed integrin that plays the same role in healthy cells.28,57 Many studies have identified the role of specific integrin subtypes in cancer progression mediated by tissue stiffness using diverse biomaterials. Using alginate–Matrigel interpenetrating networks, Chaudhuri et al. proposed a mechanism where junction α6β4 integrins and laminin play an important role in the development of malignant phenotypes in breast cancer, which is dependent on ECM stiffness. If the stiffness of the ECM is in the normal range (30 Pa), the α6β4 integrin–laminin units overlap and allow for hemidesmosome formation. However, high ECM stiffness (310 Pa) reduces hemidesmosome formation, and the free β4 can lead to Rac1 signaling and PI3 K activation for phosphorylation by receptor tyrosine kinases.14 Moreover, it was shown using 2D tunable collagen I-coated polyacrylamide (PA) hydrogels that an increase in stiffness and availability of collagen binding sites leads to the upregulation of integrin β1 in hepatocellular carcinoma (HCC) that promotes cancer progression by the activation of transforming growth factor beta-1 (TGFβ−1).98

BIOMIMETIC TUMOR MICROENVIRONMENTS

To study various organs and diseases, biomimetic environments have typically been designed to provide structure as a scaffold for cell growth and adhesion. More recently, many emerging biomaterials have been used to evaluate EMT, cell–ECM interactions, and immune cell infiltration, including organoids, decellularized ECM, natural materials, and synthetic materials. These materials and applications are summarized in Table 2.

TABLE 2.

Common biomimetic materials and their applications.

Biomaterial Cancer Application
Organoids
 Mammary epithelial cells Breast Normal tissue radiation effects on tumor-stromal and immune interactions39
 Pancreatic cancer cells Pancreatic Tumor-stromal and tumor-immune interactions129
Decellularized ECM
 Bladder-derived Skin, colon, breast Tumor growth delay and immune signatures137,147
 Mammary fat pad-derived Breast Radiation effects on tumor cell behavior, breast cancer proliferation, and invasion3
 Colorectal cancer-derived Colon Establishing the colorectal tumor microenvironment influence on macrophage-mediated cell invasion100,101
 Breast cancer-derived Breast Evaluation of tumor development and progression49,61,77,91
 Liver-derived Liver Determining how the mechanical properties of cirrhotic liver tissue alter tumor cell behavior81
 Human tumor-derived Colon Studying the mechanisms of chemotherapy resistance at different stages of malignancy48,50
 Brain tissue-derived Brain Investigating glioblastoma cell invasion66
 Colon tumor-derived Colon Evaluating tumor progression at different stages of tumorigenesis109
 Xenograft-derived Breast, liver, lung Studying the behavior of cancer cells in crosslinked dECM79
Natural materials
 HA hydrogel Breast Stiffness effects on metastasis to the brain94
 Collagen I, Matrigel Breast Recruitment of immune cells and their effect on the invasiveness of cancer cells63
 Porous chitosan-alginate scaffolds Prostate, breast, liver, brain Evaluating cancer cell behavior on scaffolds mechanically mimicking cancer progression; investigating cancer stem cell enrichment32,138
 Alginate-Matrigel Breast Effect of stiffness and ligand ratio on cell morphology and invasion11,12
 Alginate-based Lung, breast, pancreatic, neuroblastoma, pituitary Co-culture model for a drug screening platform22,27,46,71,92
Synthetic materials
 PEG-b-poly(l-alanine) hydrogels Breast Linking chemical and mechanical ECM characteristics to tumor cell behavior118
 PA hydrogels Thyroid, renal, breast Stiffness and topography effects on normal and cancer cells86,106,107
 Alginate-PCL nanofibers Liver Cancer stem cell enrichment51

Organoid and Spheroid Models

Organoid and spheroid systems are useful models for biomimetic microenvironments. Organoids are organ-like structures and are derived from cells and tissues ranging from murine to human species (Fig. 3). Organoids recapitulate key structural and functional components of various sites of interest from the brain to retinal systems.17,31,110 Organoids have been used extensively in studying multiple cancer models.8,13,24,111 For example, CRISPR/Cas9 gene editing was performed to introduce KRAS, SMAD4, APC, and TP53 mutations into normal human intestinal organoids to form adenocarcinomas in mouse models during xenograft transplantation.25 Organoid models have been developed to mimic drug resistant tumors and to evaluate therapeutic efficacy.4,17,26 Sun et al. reported the formation of organoids from reprogrammed hepatocytes to initiate HCC through c-Myc expression.120 It was shown that these organoid models can form tumors in vivo, making them a useful system for generating mouse xenografts to study drug response. In addition to tumor systems, organoids have been used as normal tissue models to evaluate tissue development as well as to study how normal tissues interact with tumor and immune cells.39,54 For example, Hacker et al. derived mammary epithelial organoids from irradiated and normal murine mammary glands.39 These organoids are being used to study the effects of normal tissue radiation damage on breast tumor-stromal and immune interactions, which may elucidate cancer recurrence mechanisms. Patient--derived organoids are also being explored to develop platforms for studying novel biomarkers and drug targets as well as tumor-immune and tumor-stromal interactions.95,104,113,127,129

FIGURE 3.

FIGURE 3.

Normal tissue and tumor organoid model systems and uses. Tumor organoid model systems can be produced from tumors or by genetic modification of organoids generated from normal tissues. Organoids can be used to answer questions about cancer biology, cell–cell interaction studies, and therapy response mechanisms.130

An additional approach to studying 3D tumor models is developing spheroids or micro-sized cell aggregates that function as in vitro models of the behavior of different tumor types.18 Limitations in the use of tumor spheroids include the lack of ECM interactions and heterogeneity in the size and shape of the aggregates. To address that variability, Pradhan et al. designed and modeled 3D tumor microspheres formed by encapsulating MCF7 breast cancer cells in a PEG-fibrinogen hydrogel. This new model presented more cancer hallmarks compared to classic spheroids, such as increased disorganization, loss in apicobasal polarity, elevated nuclear-cytoplasmic ratio, nuclear volume density, and the reduction of cell–cell junction length.102 Recently, alginate-based bioinks have been used to form 3D tissue spheroids of bone, cartilage, vascular tissue, and diverse tumors through bioprinting.5,22,37,71,92,132 Compared to previous spheroid formation techniques, this method has been shown to improve cell viability, function, and architecture. 3D printing can form a ready-to-use tumor model that can mimic interactions with the ECM and with other cells in a co-culture system. Additionally, compared to self-assembled spheroids using Matrigel, this method does not require complex components such as binding sites, growth factors, diverse extracellular fibers to promote tumor formation.122 In their work, Swaminathan et al. not only studied the pre-spheroid formation of the MDA-MB-231 and MCF7 human breast cancer cells lines but also a non-tumorigenic MCF10A breast epithelial cell line. They performed a co-culture with vascular endothelial cells to examine drug response using Matrigel, gelatin–alginate, and collagen–alginate as bioinks. Laminin was found to be a key factor in the spheroid formation of breast epithelial cells, the spheroid structure was distinct when comparing tumorigenic and non-tumorigenic cell lines, and these morphologies were conserved post-bioprinting.122

Decellularized ECM (dECM)

Decellularization is a process that removes cellular material from tissues while leaving the majority of the ECM structure intact.114 Since the 1990s, decellularized ECM (dECM) has been used for wound healing and regeneration but has been variable in its success.47 dECM can be reconstituted into hydrogels and used for in vitro and in vivo experiments to study cancer progression (Fig. 4). These dECMs consist of structural and functional molecules that assist in the 3D organization of encapsulated cells. The organs from which these matrices are derived can produce distinct ECM components, and these differences alter cell–ECM interactions that serve to influence tumor cell proliferation and adhesion.6 dECM derived from tumors has been shown to promote angiogenesis, the EMT response, and MMP-9 production.48 Many studies use ECM obtained from tumor tissue to establish the effect of the microenvironment on the behavior and progression of various types of cancer, including colorectal,48,50,100,101,109 breast,49,61,77,91 liver,81 brain,66 and lung.79 This approach allows for the evaluation of dECM obtained at different stages of tumorigenesis109 and cancer cell interactions with immune cells.100,101 One of the most important advantages of dECM is recapitulating the ECM microenvironment of specific tissues. For example, murine mammary fat pads were decellularized and exposed to radiation ex vivo.3 Using these dECM hydrogels allowed for studying the effect of radiation on breast cancer proliferation and invasion in the context of recurrence after therapy. Wolf and coworkers also showed that the ECM derived from urinary bladder can support 3T3 fibroblast growth and proliferation.136 Another notable characteristic of dECM is that it can promote a pro-regenerative immune phenotype and thus has been considered to combat tissue loss and promote wound healing following tumor resection.137

FIGURE 4.

FIGURE 4.

The process of deriving decellularized ECM from various organs for hydrogel formation.3,41,76,78,116

Naturally-Derived Materials

Matrigel

Matrigel is a solubilized basement membrane matrix that is derived from murine Engelbreth-Holm-Swarm sarcomas, tumors rich with ECM proteins such as laminin, collagen IV, and other growth factors.65 Matrigel has mostly been used as a 3D matrix for cell growth and adhesion in culture systems due to relevant cell adhesion sites. Matrigel has also been combined with other 3D systems such as alginate hydrogels as will be described below.11,12,14,73,135 Although abundantly examined, Matrigel can vary from batch to batch, and its components are undefined. Using Matrigel is nonetheless advantageous for studying how biological, chemical, and mechanical characteristics of the ECM affect the development and progression of cancer.

Alginate

Alginic acid, or alginate, is an anionic polysaccharide derived from brown algae. It is mostly comprised of α-d-mannuronic acid and β-l-guluronic acid and can form crosslinked hydrogel networks of tunable mechanical properties through crosslinking using divalent ions such a calcium and manganese.68 Alginate lacks cell adhesion binding sites and often requires modification with peptides or combination with collagen or Matrigel to form interpenetrating networks.14 Read et al. showed that human colorectal cancer cells were viable when encapsulated within unmodified alginate hydrogels to study metabolic effects after irradiation.105 However, not having binding sites available for the cells reduces the accuracy of the biomimetic environment since the progression of cancer is connected to cell–ECM dynamics.131 In order to mitigate this, Cavo et al. developed an alginate hydrogel conjugated with RGD peptides that bind to integrins such as αvβ3 and α5β1.11 Other peptide sequences such as DGEA and YIGSR, which are involved in cell adhesion to the ECM, have been used in conjunction with alginate to further produce bio-mimetic environments. It has been shown that the expression of these peptides can control EMT.68 Alginate has also been used with collagen for immunotherapy treatments in mouse studies by Wei and coworkers. They show that the alginate–collagen matrix along with the TLR7 agonist R837 and radiation decreases 4T1 primary tumor growth and reduces the volume of distal tumors.87 Alginate-chitosan scaffolds have been designed to enhance cell adhesion. These scaffolds have been used to mimic stages of cancer progression and metastasis in prostate adenocarcinoma and to examine cancer stem-like enrichment in prostate, breast, liver, and brain cancer.32,138

Since alginate can be easily tuned through the addition of divalent cations, it is widely used for evaluating biomimetic stiffnesses of the tumor microenvironment. Stiffness is an important factor when studying the tumor microenvironment as it has been shown that matrix rigidity can cause cells to undergo EMT that is characteristic of invasive phenotypes (Fig. 5a).10,21,108 It is also important to tune biomaterials to mimic the vast range of mechanical properties experienced by cells in various organ microenvironments, including breast tissue (0.5–1 kPa), the intestines (20–40 kPa), and bone (15,000–20,000 kPa) (Fig. 5b). Alginate–Matrigel hydrogels have been used for breast cancer dormancy exit studies in relation to microenvironmental stiffness. Chaudhuri et al. fabricated alginate-Matrigel interpenetrating networks to evaluate breast cancer cell behavior in environments ranging from 30 to 300 Pa to mimic progressive tumors. Using the MCF10A breast cancer cell line, they observed that increased stiffness can promote the transition from epithelial to mesenchymal phenotypes, which is observed when dormant tumor cells exit dormancy.14 This alginate–Matrigel model has also been used to study how mechanotransduction pathways such as YAP/TAZ signaling relate to breast cancer progression.73 These models for integrin binding have also been used to study the invasiveness of tumor cells. Cavo et al. used alginate–Matrigel hydrogels to understand how stiffness can cause malignant progression as it has been shown that the biomechanical properties directly affect neoplastic disease.121 MCF-7 ER+ breast cancer cells were shown to exhibit a rounded morphology in microenvironments of 5 kPa compared to MDA-MB-231 cells, an ER-cancer cell line with higher metastatic and invasive potential.11,12

FIGURE 5.

FIGURE 5.

Biomimetic materials have been developed to span the stiffness range observed in (a) the epithelial-to-mesenchymal transition (EMT) and (b) commonly studied tissues and organs.9,19,45,108

Synthetic Materials

Polyethylene Glycol (PEG)

PEG is a synthetic polymer that is used in a variety of biomedical applications.90 PEG, when modified with methacrylate and dimethacrylate, is photocrosslinkable under UV light and can easily be conjugated with active protein sites for cell adhesion to better evaluate cell morphology and production of growth factors.55,117 This material has been used to fabricate biologically active microenvironments by conjugating PEG to different protein binding sites such as RGD, IKVAV, and GFOGER to mimic the cellular binding sites in the fibrous structure of the ECM.82,115,141 Gill and coworkers used PEG conjugated with RGDS (PEG-RGDS) for cell adhesion and GGGPQGIWGQGK (PQ) for degradation by matrix metalloproteinases. In this model, lung adenocarcinoma cells were used to understand morphological and epithelial changes in response to microenvironmental stiffness, ligand adhesion concentration, and TGFβ.34 Stiffnesses were varied from 21 to 55 kPa in order to study the effect of stiffness on the morphology of lung adenocarcinoma cells. Higher stiffnesses promoted an increase in mesenchymal phenotypes. In addition, cells interacting with larger amounts of RGDS peptide presented an epithelial phenotype. Sawicki and coworkers varied the concentration of the peptides GFOGER, RGDS, and IKVAV that bind to collagen, fibronectin, and laminin, respectively.115 These concentrations were changed to mimic peptide concentrations associated with cancerous tissue, including increased collagen I binding sites.128

Polyacrylamide (PA)

Acrylamide-based hydrogels are widely used to model the stiffness range present in biological matrices, typically from 0.1 to 119 kPa.15 PA can be functionalized and coated with ECM components such as fibronectin, vitronectin, and collagen to encourage cell adhesion.20 A major challenge, however, in using this material is the cytotoxicity of acrylamide monomers, which skews studies toward 2D models.145 Currently, PA hydrogels are being deigned to understand how mechanical cues such as stiffness, topography, and geometry alter gene expression,86 EMT, drug resistance,108 migration,69 and tumor progression.86,106,107 To evaluate mechanotransduction in gradient hydrogels, Hadden et al. fabricated PA-derived gels to create cell culture surfaces that have tunable stiffnesses.40 Using human adipose-derived stem cells, it was reported that cells experienced durotaxis on fibronectin-coated PA gels with a steep 8.2 kPa/mm gradient while cells did not migrate in response to a more shallow gradient of 2.9 kPa/mm. This indicates that conditions separating the effects of durotaxis from stiffness response should be considered in studying tumor cell behavior. PA hydrogels have also been used to understand cancer stem cell (CSC) plasticity. Tian et al. used PA hydrogels to understand whether matrix rigidity has an effect on the phenotype of CSCs.125 It was reported that HCCs had stem cell-like properties and poor spreading at 5.9 kPa corresponding to normal liver stiffness compared to stiffnesses in the range of cancerous tissues (48.1 kPa). PA surfaces have also been utilized to study the response of human primary thyroid cells (S747) and anaplastic thyroid carcinoma cells (S277) when sensing a range of mechanical environments.107 It was found that normal cells adapt their viscoelastic properties in response to stiffness changes while the carcinoma cell viscoelasticity remained constant.

Other Synthetic Biomaterials

Other commonly studied synthetic biomimetic materials are polydimethylsiloxane (PDMS), polycaprolactone (PCL), and polyurethane (PUR) films.51,75,97 All of these materials allow for a higher degree of stiffness matching of tissues and organs compared to natural biomaterials, and they can also be functionalized or mixed with ECM components to form hybrid materials that can model the tumor microenvironment.97 One notable example evaluates the development of porous and tunable alginate-PCL nanofiber scaffolds for investigating cancer stem-like cell enrichment, EMT, and cell distribution.51

IMMUNE CELL BEHAVIOR

Studying the Immunological Response in Cancer Microenvironments

There are several components of the microenvironment that influence tumor growth, including cancer-associated fibroblasts, tumor associated neutrophils, and tumor associated macrophages (TAMs).64,93 It has been shown that these cellular and immune populations, or lack thereof, can contribute to cancer progression and metastasis. For example, triple negative breast cancer tumor cell recruitment after radiation therapy was enhanced due to reduced lymphocytes and excess macrophages.103 In addition, incorporating chemokines and cytokines into models are important as they enable pro-inflammatory responses that affect the growth of tumors and give signals to immune cells to encourage malignant behavior.64,140

One overarching goal in designing 3D biomimetic systems is to understand how tumor cells evade the immune response.36 TAMs are associated with poor prognosis in patients and promote tumor growth and migration.85,147 A biomimetic materials approach has been undertaken to evaluate immune cell interactions with stromal and tumor cells. Wolf et al. used urinary bladder matrix (UBM) scaffolds embedded with three different cancer cells, melanoma (B16-F10), colorectal carcinoma (CT26), and mammary carcinoma (4T1) to evaluate how the ECM microenvironment influenced tumor progression. UBM is a decellularized scaffold composed of basal lamina and lamina propria of the porcine urinary bladder that contains collagen I, ECM-associated factors, glycoproteins, and proteoglycans.112 The UBM scaffold inhibited and delayed tumor formation while promoting a distinct immune signature that was dependent on CD4+ T cell and macrophage infiltration.137 In addition, Zhu and co-authors have shown that cellular phenotypes and their responses can change within dECM hydrogels. dECM hydrogels were found to play a role in macrophage activation and polarization that was dependent on the organ from which the ECM was derived.147 Keane et al. demonstrated that decellularized matrices can promote anti-inflammatory responses by reducing inflammatory macrophage infiltration.62 The mechanisms behind how dECM can activate these macro-phages warrant further study in order to evaluate polarization that mimics the tumor microenvironment.

3D bioprinted tumor models have also been used to study the interactions between different cell types, including immune, cancer, endothelial, and fibroblast cells in vitro.46,71 Gelatin and gelatin methacrylate have been used for this application.89 In their work, Heinrich et al. fabricated a mini-brain with encapsulated glioblastoma-associated macrophages (GAMs) and glioblastoma multiforme (GBM) tumor cells to study cell crosstalk in vitro. This work demonstrated how the presence of GAMs promotes the invasion and proliferation of GBM as well as how GBM leads to GAM recruitment and proliferation.46

An important application of biomimetic materials is to study the behavior of cancer cells treated with novel therapies.3 One approach is local cancer immunotherapy, a treatment intended to stimulate the immune system of cancer patients commonly achieved through T-cell infusion, vaccines, or antibodies to inhibit proteins produced by cancer cells using macroscale bio-materials.29,83 The principal advantage of local immunotherapy is specific immunomodulation at the tumor site, which prevents systemic toxicity. Injectable biomaterials can include cargo such as immunomodulators, immune cells, or cancer vaccines. Mesoporous silica microrods, PEG with polypeptide blocks, synthetic peptides with specific motifs, and DNA-scaffolded biomaterials are being developed for this purpose.53,56,74,142 Additionally, natural biopolymers such as alginate, chitosan, gelatin, and HA have been explored.74,139 Lee et al. used DNA polyaptamer hydrogels with Cas9/sgRNA to promote the controlled delivery of immune checkpoint inhibitors for cancer immunotherapy. The hydrogels are composed of two templates that contain PD-1 aptamers and prepared by rolling circle amplification containing complementary sequences of sgRNA to be cleaved by Cas9. The release of a programmed death receptor (PD-1) DNA aptamer promotes the activation of T-cells and reduces tumor growth.72 Furthermore, immune checkpoint inhibitors can be released with a hydrogel-based polypeptide vaccine to improve tumor immunotherapy. Song et al. developed an injectable PEG-b-poly(l-alanine) hydrogel with encapsulated granulocyte–macrophage colony-stimulating factor and anti-CTLA-4/PD-1 antibodies. The injectable hydrogel promoted the recruitment and activation of dendritic and T-cells. Moreover, the hydrogel vaccine led to the secretion of specific cytokines that reduced the growth of B16 melanoma tumors. These injectable biomaterials can be used as a sustained delivery platform with co-delivery of immunotherapeutics to promote immune cell activation and eliminate tumors.118

Cell Membrane-Mimicking Nanoparticles

Currently, one of the major limitations in the use of drug delivery nanoparticles in chemotherapy is low therapeutic efficacy and limited tumor penetration.59 For that reason, developing biomimetic nanoparticles as drug delivery vehicles to promote an immunomodulatory response has been explored. These nanoparticles are composed of a cell membrane coating and can mimic specific cell types due to the surface functionalization of the originating cell’s membrane proteins.16,23,60,67,119,126,143,146 An innovative system for targeting tumors is the design of paclitaxel-loaded polymeric nanoparticles coated with the cell membrane of a specific tumor cell. For example, Sun et al. fabricated cancer cell biomimetic nanoparticles from 4T1 murine triple negative breast cancer cell membrane-derived vesicles and paclitaxel-loaded polymeric nanoparticles composed of PCL. In contrast with other drug delivery systems, this class of biomimetic nanoparticle has a high specificity for targeting primary tumors.119 In conjunction with other materials, PEG has also been used to help create immunological responses in a drug delivery system. Lai and coworkers fabricated DSPE-PEG nanoparticles coated with macrophage plasma membranes as a functional macrophage mimic.70 This approach showed minimal cytotoxicity and successfully allowed the nanoparticles to cross the blood brain barrier to selectively accumulate at glioblastoma sites, making this an optimal delivery strategy for reducing tumor growth.147

CONCLUSION

The development of biomimetic materials offers promising avenues of understanding tumor and immune cell behavior in vitro and in vivo. In this review, we have demonstrated how various materials and techniques are used to evaluate the phenotypic and functional changes in tumor and immune cells. Models composed of common biomaterials such as alginate and PEG, novel materials including DNA hydrogels and dECM, and new technologies like 3D bioprinting allow for recapitulating the complex tumor microenvironment and analyzing multiple variables that promote tumor progression.

Biomimetic materials and synthetic environments are being designed in order to replicate complex microenvironments of interest and answer relevant biological questions. Some biomimetic models, such as organoids, are being implanted in vivo to encourage a vascularized environment.84 Other studies have also begun to incorporate hydrogels into bioreactors to better mimic physiological conditions.38,88,96 Despite the development of these biomimetic materials and new, innovative ways of using them, not all of the factors that influence tumor progression can be tested simultaneously. This necessitates the continued use of in vivo models to validate in vitro observations. The complexity of design and how closely these models will resemble living systems remains to be seen. Nonetheless, these biomimetic materials allow for the study of tumor cell crosstalk with immune cells, mechanotransduction, ligand density, and cell–ECM interactions, which can advance the development of more effective therapies leading to improved patient outcomes.

ACKNOWLEDGMENTS

The authors gratefully acknowledge financial support from the National Institutes of Health and the National Cancer Institute [Grants #R00CA201304, #T32CA119925 (LAN)].

Footnotes

Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

REFERENCES

  • 1.Akhavan A, Griffith OL, Soroceanu L, Leonoudakis D, Luciani-Torres MG, Daemen A, Gray JW, and Muschler JL. Loss of cell-surface laminin anchoring promotes tumor growth and is associated with poor clinical outcomes. Cancer Res 72:2578–2588, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Aljghami ME, Saboor S, and Amini-Nik S. Emerging innovative wound dressings. Ann. Biomed. Eng 47:659–675, 2019. [DOI] [PubMed] [Google Scholar]
  • 3.Alves SM, Zhu T, Shostak A, Rossen NS, and Rafat M. Studying normal tissue radiation effects using extracellular matrix hydrogels. J. Vis. Exp 149:e59304, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Astashkina A, and Grainger DW. Critical analysis of 3-D organoid in vitro cell culture models for high-throughput drug candidate toxicity assessments. Adv. Drug Deliv. Rev 69–70:1–18, 2014. [DOI] [PubMed] [Google Scholar]
  • 5.Axpe E, and Oyen M. Applications of alginate-based bioinks in 3D bioprinting. Int. J. Mol. Sci 17:1976, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Beachley VZ, Wolf MT, Sadtler K, Manda SS, Jacobs H, Blatchley MR, Bader JS, Pandey A, Pardoll D, and Elisseeff JH. Tissue matrix arrays for high-throughput screening and systems analysis of cell function. Nat. Methods 12:1197–1204, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bissell MJ, and Radisky D. Putting tumours in context. Nat. Rev. Cancer 1:46–54, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bleijs M, Wetering M, Clevers H, and Drost J. Xenograft and organoid model systems in cancer research. EMBO J 38:1–11, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Booth AJ, Hadley R, Cornett AM, Dreffs AA, Matthes SA, Tsui JL, Weiss K, Horowitz JC, Fiore VF, Barker TH, Moore BB, Martinez FJ, Niklason LE, and White ES. Acellular normal and fibrotic human lung matrices as a culture system for in vitro investigation. Am. J. Respir. Crit. Care Med 186:866–876, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Brabletz T, Kalluri R, Nieto MA, and Weinberg RA. EMT in cancer. Nat. Rev. Cancer 18:128–134, 2018. [DOI] [PubMed] [Google Scholar]
  • 11.Cavo M, Caria M, Pulsoni I, Beltrame F, Fato M, and Scaglione S. A new cell-laden 3D Alginate-Matrigel hydrogel resembles human breast cancer cell malignant morphology, spread and invasion capability observed ‘‘in vivo’’. Sci. Rep 8:1–12, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cavo M, Fato M, Peñuela L, Beltrame F, Raiteri R, and Scaglione S. Microenvironment complexity and matrix stiffness regulate breast cancer cell activity in a 3D in vitro model. Sci. Rep 6:35367, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chan AS, Yan HHN, and Leung SY. Break-through moments: organoid models of cancer. Cell Stem Cell 24:839–840, 2019. [DOI] [PubMed] [Google Scholar]
  • 14.Chaudhuri O, Koshy ST, Da Cunha CB, Shin JW, Verbeke CS, Allison KH, and Mooney DJ. Extra-cellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater 13:970–978, 2014. [DOI] [PubMed] [Google Scholar]
  • 15.Chaudhuri PK, Low BC, and Lim CT. Mechanobiology of tumor growth. Chem. Rev 118:6499–6515, 2018. [DOI] [PubMed] [Google Scholar]
  • 16.Chen Z, Zhao P, Luo Z, Zheng M, Tian H, Gong P, Gao G, Pan H, Liu L, Ma A, Cui H, Ma Y, and Cai L. Cancer cell membrane-biomimetic nanoparticles for homologous-targeting dual-modal imaging and photothermal therapy. ACS Nano 10:10049–10057, 2016. [DOI] [PubMed] [Google Scholar]
  • 17.Clevers H Modeling development and disease with organoids. Cell 165:1586–1597, 2016. [DOI] [PubMed] [Google Scholar]
  • 18.Costa EC, Moreira AF, de Melo-Diogo D, Gaspar VM, Carvalho MP, and Correia IJ. 3D tumor spheroids: an overview on the tools and techniques used for their analysis. Biotechnol. Adv 34:1427–1441, 2016. [DOI] [PubMed] [Google Scholar]
  • 19.Cox TR, and Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis. Model. Mech 4:165–178, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cretu A, Castagnino P, and Assoian R. Studying the effects of matrix stiffness on cellular function using acrylamide-based hydrogels. J. Vis. Exp 42:e2089, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dai J, Qin L, Chen Y, Wang H, Lin G, Li X, Liao H, and Fang H. Matrix stiffness regulates epithelial-mesenchymal transition via cytoskeletal remodeling and MRTF-A translocation in osteosarcoma cells. J. Mech. Behav. Biomed. Mater 90:226–238, 2019. [DOI] [PubMed] [Google Scholar]
  • 22.Diao J, Zhang C, Zhang D, Wang X, Zhang J, Ma C, Deng K, Jiang T, Jia W, and Xu T. Role and mechanisms of a three-dimensional bioprinted microtissue model in promoting proliferation and invasion of growth-hormone-secreting pituitary adenoma cells. Biofabrication 11:025006, 2019. [DOI] [PubMed] [Google Scholar]
  • 23.Díaz-Saldívar P, and Huidobro-Toro JP. ATP-loaded biomimetic nanoparticles as controlled release system for extracellular drugs in cancer applications. Int. J. Nanomed 14:2433–2447, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Drost J, and Clevers H. Organoids in cancer research. Nat. Rev. Cancer 18:407–418, 2018. [DOI] [PubMed] [Google Scholar]
  • 25.Drost J, Van Jaarsveld RH, Ponsioen B, Zimberlin C, Van Boxtel R, Buijs A, Sachs N, Overmeer RM, Offerhaus GJ, Begthel H, Korving J, Van De Wetering M, Schwank G, Logtenberg M, Cuppen E, Snippert HJ, Medema JP, Kops GJPL, and Clevers H. Sequential cancer mutations in cultured human intestinal stem cells. Nature 521:43–47, 2015. [DOI] [PubMed] [Google Scholar]
  • 26.Duarte AA, Gogola E, Sachs N, Barazas M, Annunziato S, De Ruiter JR, Velds A, Blatter S, Houthuijzen JM, Van De Ven M, Clevers H, Borst P, Jonkers J, and Rottenberg S. BRCA-deficient mouse mammary tumor organoids to study cancer-drug resistance. Nat. Methods 15:134–140, 2018. [DOI] [PubMed] [Google Scholar]
  • 27.Duarte Campos DF, Bonnin Marquez A, O’Seanain C, Fischer H, Blaeser A, Vogt M, Corallo D, and Aveic S. Exploring cancer cell behavior in vitro in threedimensional multicellular bioprintable collagen-based hydrogels. Cancers 11:180, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Elosegui-Artola A, Bazelliéres E, Allen MD, Andreu I, Oria R, Sunyer R, Gomm JJ, Marshall JF, Jones JL, Trepat X, and Roca-Cusachs P. Rigidity sensing and adaptation through regulation of integrin types. Nat. Mater 13:631–637, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Eskiizmir G, Baskın Y, and Yapıcı K. Graphene-based nanomaterials in cancer treatment and diagnosis. Fullerens, Graphenes Nanotubes, pp. 331–374, 2018. [Google Scholar]
  • 30.Fang M, Yuan J, Peng C, and Li Y. Collagen as a double-edged sword in tumor progression. Tumour Biol 35:2871–2882, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fatehullah A, Tan SH, and Barker N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol 18:246–254, 2016. [DOI] [PubMed] [Google Scholar]
  • 32.Florczyk SJ, Kievit FM, Wang K, Erickson AE, Ellenbogen RG, and Zhang M. 3D porous chitosan–alginate scaffolds promote proliferation and enrichment of cancer stem-like cells. J. Mater. Chem. B 4:6326–6334, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Frantz C, Stewart KM, and Weaver VM. The extracellular matrix at a glance. J. Cell Sci 123:4195–4200, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gill BJ, Gibbons DL, Roudsari LC, Saik JE, Rizvi ZH, Roybal JD, Kurie JM, and West JL. A synthetic matrix with independently tunable biochemistry and mechanical properties to study epithelial morphogenesis and EMT in a lung adenocarcinoma model. Cancer Res 72:6013–6023, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Glaser DE, and Viney C. Biomimetic Materials. Biomaterial Science, pp. 349–360, 2013. [Google Scholar]
  • 36.Gonzalez H, Hagerling C, and Werb Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev 32:1267–1284, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gopinathan J, and Noh I. Recent trends in bioinks for 3D printing. Biomater. Res 22:11, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Guller AE, Grebenyuk PN, Shekhter AB, Zvyagin AV, and Deyev SM. Bioreactor-based tumor tissue engineering. Acta Nat 8:44–58, 2016. [PMC free article] [PubMed] [Google Scholar]
  • 39.Hacker BC, Gomez JD, Batista CAS, and Rafat M. Growth and characterization of irradiated organoids from mammary glands. J. Vis. Exp 147:e59293, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hadden WJ, Young JL, Holle AW, McFetridge ML, Kim DY, Wijesinghe P, Taylor-Weiner H, Wen JH, Lee AR, Bieback K, Vo BN, Sampson DD, Kennedy BF, Spatz JP, Engler AJ, and Cho YS. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. Proc. Natl. Acad. Sci. USA 114:5647–5652, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Haggerty AE, and Oudega M. Biomaterials for spinal cord repair. Neurosci. Bull 29:445–459, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hamidi H, and Ivaska J. Every step of the way: integrins in cancer progression and metastasis. Nat Rev. Can 18:533–548, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Han Z, and Lu ZR. Targeting fibronectin for cancer imaging and therapy. J. Mater. Chem. B 5:639–654, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hanahan D, and Weinberg RA. Hallmarks of cancer: the next generation. Cell 144:646–674, 2011. [DOI] [PubMed] [Google Scholar]
  • 45.Handorf AM, Zhou Y, Halanski MA, and Li W-J. Tissue stiffness dictates development, homeostasis, and disease progression. Organogenesis 11:1–15, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Heinrich MA, Bansal R, Lammers T, Zhang YS, Michel Schiffelers R, and Prakash J. 3D-bioprinted minibrain: a glioblastoma model to study cellular interactions and therapeutics. Adv. Mater 31:1806590, 2019. [DOI] [PubMed] [Google Scholar]
  • 47.Hinderer S, Layland SL, and Schenke-Layland K. ECM and ECM-like materials: biomaterials for applications in regenerative medicine and cancer therapy. Adv. Drug Deliv. Rev 97:260–269, 2016. [DOI] [PubMed] [Google Scholar]
  • 48.Hoshiba T Decellularized extracellular matrix for cancer research. Materials 12:1–16, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hoshiba T, and Tanaka M. Breast cancer cell behaviors on staged tumorigenesis-mimicking matrices derived from tumor cells at various malignant stages. Biochem. Biophys. Res. Commun 439:291–296, 2013. [DOI] [PubMed] [Google Scholar]
  • 50.Hoshiba T, and Tanaka M. Decellularized matrices as in vitro models of extracellular matrix in tumor tissues at different malignant levels: mechanism of 5-fluorouracil resistance in colorectal tumor cells. Biochim. Biophys. Acta Mol. Cell Res 2749–2757:2016, 1863. [DOI] [PubMed] [Google Scholar]
  • 51.Hu W-W, Lin C-H, and Hong Z-J. The enrichment of cancer stem cells using composite alginate/polycaprolactone nanofibers. Carbohydr. Polym 206:70–79, 2019. [DOI] [PubMed] [Google Scholar]
  • 52.Huang S, and Ingber DE. Cell tension, matrix mechanics, and cancer development. Cancer Cell 8:175–176, 2005. [DOI] [PubMed] [Google Scholar]
  • 53.Huang X, Williams JZ, Chang R, Li Z, Gai E, Patterson DM, Wei Y, Lim WA, and Desai TA. DNA-scaffolded biomaterials enable modular and tunable control of cell-based cancer immunotherapies. bioRxiv 1:587105, 2019. [Google Scholar]
  • 54.Huch M, and Koo B-K. Modeling mouse and human development using organoid cultures. Development 142:3113–3125, 2015. [DOI] [PubMed] [Google Scholar]
  • 55.Hwang JW, Noh SM, Kim B, and Jung HW. Gelation and crosslinking characteristics of photopolymerized poly(ethylene glycol) hydrogels. J. Appl. Polym. Sci 132:1–6, 2015.25866416 [Google Scholar]
  • 56.Ishii S, Kaneko J, and Nagasaki Y. Development of a long-acting, protein-loaded, redox-active, injectable gel formed by a polyion complex for local protein therapeutics. Biomaterials 84:210–218, 2016. [DOI] [PubMed] [Google Scholar]
  • 57.Jang I, Beningo KA, Jang I, and Beningo KA. Integrins, CAFs and mechanical forces in the progression of cancer. Cancers 11:721, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jin H, and Varner J. Integrins: roles in cancer development and as treatment targets. Br. J. Cancer 90:561–565, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Jin H, Zhu T, Huang X, Sun M, Li H, Zhu X, Liu M, Xie Y, Huang W, and Yan D. ROS-responsive nanoparticles based on amphiphilic hyperbranched polyphosphoester for drug delivery: light-triggered size-reducing and enhanced tumor penetration. Biomaterials 211:68–80, 2019. [DOI] [PubMed] [Google Scholar]
  • 60.Jin J, Krishnamachary B, Barnett JD, Chatterjee S, Chang D, Mironchik Y, Wildes F, Jaffee EM, Nimmagadda S, and Bhujwalla ZM. Human cancer cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated invasion and metastasis and induce T-cells. ACS Appl. Mater. Interfaces 11:7850–7861, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jin Q, Liu G, Li S, Yuan H, Yun Z, Zhang W, Zhang S, Dai Y, and Ma Y. Decellularized breast matrix as bioactive microenvironment for in vitro three-dimensional cancer culture. J. Cell. Physiol 234:3425–3435, 2019. [DOI] [PubMed] [Google Scholar]
  • 62.Keane TJ, Dziki J, Sobieski E, Smoulder A, Castleton A, Turner N, White LJ, and Badylak SF. Restoring mucosal barrier function and modifying macrophage phenotype with an extracellular matrix hydrogel: potential therapy for ulcerative colitis. J. Crohns. Colitis 11:360–368, 2017. [DOI] [PubMed] [Google Scholar]
  • 63.Kim H, Chung H, Kim J, Choi D, Shin Y, Kang YG, Kim B, Seo S, Chung S, and Seok SH. Macrophages-triggered sequential remodeling of endothelium-interstitial matrix to form pre-metastatic niche in microfluidic tumor microenvironment. Adv. Sci 6:1900195, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kim J, and Bae JS. Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm 2016, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kleinman HK, and Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 15:378–386, 2005. [DOI] [PubMed] [Google Scholar]
  • 66.Koh I, Cha J, Park J, Choi J, Kang S-G, and Kim P. The mode and dynamics of glioblastoma cell invasion into a decellularized tissue-derived extracellular matrix-based three-dimensional tumor model. Sci. Rep 8:4608, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kroll AV, Jiang Y, Zhou J, Holay M, Fang RH, and Zhang L. Biomimetic nanoparticle vaccines for cancer therapy. Adv. Biosyst 3:1800219, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kuen YL, and Mooney DJ. Alginate: properties and biomedical applications. Prog. Polym. Sci 37:106–126, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kuo C-HR, Xian J, Brenton JD, Franze K, and Sivaniah E. Complex stiffness gradient substrates for studying mechanotactic cell migration. Adv. Mater 24:6059–6064, 2012. [DOI] [PubMed] [Google Scholar]
  • 70.Lai J, Deng G, Sun Z, Peng X, Li J, Gong P, Zhang P, and Cai L. Scaffolds biomimicking macrophages for a glioblastoma NIR-Ib imaging guided photothermal therapeutic strategy by crossing Blood-Brain Barrier. Biomaterials 211:48–56, 2019. [DOI] [PubMed] [Google Scholar]
  • 71.Langer EM, Allen-Petersen BL, King SM, Kendsersky ND, Turnidge MA, Kuziel GM, Riggers R, Samatham R, Amery TS, Jacques SL, Sheppard BC, Korkola JE, Muschler JL, Thibault G, Chang YH, Gray JW, Presnell SC, Nguyen DG, and Sears RC. Modeling tumor phenotypes in vitro with threedimensional bioprinting. Cell Rep 26:608.e6–623.e6, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lee J, Le Q-V, Yang G, and Oh Y-K. Cas9-edited immune checkpoint blockade PD-1 DNA polyaptamer hydrogel for cancer immunotherapy. Biomaterials 218:119359, 2019. [DOI] [PubMed] [Google Scholar]
  • 73.Lee JY, Chang J, Dominguez AA, Nam S, Chang J, Lee H, Varma S, Qi LS, West RB, and Chaudhuri O. YAP-independent mechanotransduction drives breast cancer progression. bioRxiv 1:495499, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lei K, and Tang L. Surgery-free injectable macroscale biomaterials for local cancer immunotherapy. Biomater. Sci 7:733–749, 2019. [DOI] [PubMed] [Google Scholar]
  • 75.Lemma ED, Sergio S, Spagnolo B, Pisanello M, Algieri L, Coluccia MA, Maffia M, De Vittorio M, and Pisanello F. Tunable mechanical properties of stent-like microscaffolds for studying cancer cell recognition of stiffness gradients. Microelectron. Eng 190:11–18, 2018. [Google Scholar]
  • 76.Lewis PL, Su J, Yan M, Meng F, Glaser SS, Alpini GD, Green RM, Sosa-Pineda B, and Shah RN. Complex bile duct network formation within liver decellularized extracellular matrix hydrogels. Sci. Rep 8:12220, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu G, Wang B, Li S, Jin Q, and Dai Y. Human breast cancer decellularized scaffolds promote epithelial-to-mesenchymal transitions and stemness of breast cancer cells in vitro. J. Cell. Physiol 234:9447–9456, 2019. [DOI] [PubMed] [Google Scholar]
  • 78.Lopresti ST, and Brown BN. Host response to naturally derived biomaterials. Host Response to Biomaterial, pp. 53–79, 2015. [Google Scholar]
  • 79.Lü WD, Sun RF, Hu YR, Lu JR, Gu L, Liu ZG, Lei GY, Qiang Z, and Cai L. Photooxidatively crosslinked acellular tumor extracellular matrices as potential tumor engineering scaffolds. Acta Biomater 71:460–473, 2018. [DOI] [PubMed] [Google Scholar]
  • 80.Luca AC, Mersch S, Deenen R, Schmidt S, Messner I, Schäfer KL, Baldus SE, Huckenbeck W, Piekorz RP, Knoefel WT, Krieg A, and Stoecklein NH. Impact of the 3D microenvironment on phenotype, gene expression, and EGFR inhibition of colorectal cancer cell lines. PLoS ONE 8:e59689, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ma X, Yu C, Wang P, Xu W, Wan X, Lai CSE, Liu J, Koroleva-Maharajh A, and Chen S. Rapid 3D bioprinting of decellularized extracellular matrix with regionally varied mechanical properties and biomimetic microarchitecture. Biomaterials 185:310–321, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Macdougall LJ, Wiley KL, Kloxin AM, and Dove AP. Design of synthetic extracellular matrices for probing breast cancer cell growth using robust cycto-compatible nucleophilic thiol-yne addition chemistry. Biomaterials 178:435–447, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Manmohan S, and Salnikova M. Novel Approaches and Strategies for Biologics, Vaccines and Cancer Therapies New York: Academic Press, 2015. [Google Scholar]
  • 84.Mansour AA, Gonçalves JT, Bloyd CW, Li H, Fernandes S, Quang D, Johnston S, Parylak SL, Jin X, and Gage FH. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol 36:432–441, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mantovani A, Marchesi F, Malesci A, Laghi L, and Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol 14:399–416, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Medina SH, Bush B, Cam M, Sevcik E, Del-Rio FW, Nandy K, and Schneider JP. Identification of a mechanogenetic link between substrate stiffness and chemotherapeutic response in breast cancer. Biomaterials 202:1–11, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mei E, Li S, Song J, Xing R, Li Z, and Yan X. Self-assembling collagen/alginate hybrid hydrogels for combinatorial photothermal and immuno tumor therapy. Colloids Surf. A 577:570–575, 2019. [Google Scholar]
  • 88.Meinert C, Schrobback K, Hutmacher DW, and Klein TJ. A novel bioreactor system for biaxial mechanical loading enhances the properties of tissue-engineered human cartilage. Sci. Rep 7:1–14, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Meng F, Meyer CM, Joung D, Vallera DA, McAlpine MC, and Panoskaltsis-Mortari A. 3D bioprinted in vitro metastatic models via reconstruction of tumor microenvironments. Adv. Mater 31:1806899, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Migonney V History of Biomaterials. Biomaterials, pp. 1–10, 2014. [Google Scholar]
  • 91.Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer X-L, Sachs PC, and Bruno RD. 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomater 95:201–213, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Mondal A, Gebeyehu A, Subramanian R, Rishi A, and Singh M. Abstract 5018: bioprinted (3D) co-cultured spheroids with NSCLC PDX cells and cancer associated fibroblasts (CAFs) using alginate/gelatin hydrogel. Cancer Res 78(13 Suppl):2018, 2018. [Google Scholar]
  • 93.Monteran L, and Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immuno-suppression in the tumor microenvironment. Front. Immunol 10:1–15, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Narkhede AA, Crenshaw JH, Manning RM, and Rao SS. The influence of matrix stiffness on the behavior of brain metastatic breast cancer cells in a biomimetic hyaluronic acid hydrogel platform. J. Biomed. Mater. Res. A 106:1832–1841, 2018. [DOI] [PubMed] [Google Scholar]
  • 95.Neal JT, et al. Organoid modeling of the tumor immune microenvironment. Cell 175:1972.e16–1988.e16, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Novak CM, Horst EN, Taylor CC, Liu CZ, and Mehta G. Fluid shear stress stimulates breast cancer cells to display invasive and chemoresistant phenotypes while upregulating PLAU in a 3D bioreactor. Biotechnol. Bioeng, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Page JM, Merkel AR, Ruppender NS, Guo R, Dadwal UC, Cannonier SA, Basu S, Guelcher SA, and Sterling JA. Matrix rigidity regulates the transition of tumor cells to a bone-destructive phenotype through integrin β3 and TGF-β receptor type II. Biomaterials 64:33–44, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pang M, Teng Y, Huang J, Yuan Y, Lin F, and Xiong C. Substrate stiffness promotes latent TGF-β1 activation in hepatocellular carcinoma. Biochem. Biophys. Res. Commun 483:553–558, 2017. [DOI] [PubMed] [Google Scholar]
  • 99.Pfeifer CR, Alvey CM, Irianto J, and Discher DE. Genome variation across cancers scales with tissue stiffness: an invasion-mutation mechanism and implications for immune cell infiltration. Curr. Opin. Syst. Biol 2:103–114, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Piccoli M, D’Angelo E, Crotti S, Sensi F, Urbani L, Maghin E, Burns A, De Coppi P, Fassan M, Rugge M, Rizzolio F, Giordano A, Pilati P, Mammano E, Pucciarelli S, and Agostini M. Decellularized colorectal cancer matrix as bioactive microenvironment for in vitro 3D cancer research. J. Cell. Physiol 233:5937–5948, 2018. [DOI] [PubMed] [Google Scholar]
  • 101.Pinto ML, Rios E, Silva AC, Neves SC, Caires HR, Pinto AT, Durães C, Carvalho FA, Cardoso AP, Santos NC, Barrias CC, Nascimento DS, Pinto-do-Ó P, Barbosa MA, Carneiro F, and Oliveira MJ. Decellularized human colorectal cancer matrices polarize macrophages towards an anti-inflammatory phenotype promoting cancer cell invasion via CCL18. Biomaterials 124:211–224, 2017. [DOI] [PubMed] [Google Scholar]
  • 102.Pradhan S, Clary JM, Seliktar D, and Lipke EA. A three-dimensional spheroidal cancer model based on PEG-fibrinogen hydrogel microspheres. Biomaterials 115:141–154, 2017. [DOI] [PubMed] [Google Scholar]
  • 103.Rafat M, Aguilera TA, Vilalta M, Bronsart LL, Soto LA, Von Eyben R, Golla MA, Ahrari Y, Melemenidis S, Afghahi A, Jenkins MJ, Kurian AW, Horst KC, Giaccia AJ, and Graves EE. Macrophages promote circulating tumor cell-mediated local recurrence following radiotherapy in immunosuppressed patients. Cancer Res 78:4241–4252, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ramamoorthy P, Thomas SM, Kaushik G, Subramaniam D, Chastain KM, Dhar A, Tawfik O, Kasi A, Sun W, Ramalingam S, Gunewardena S, Umar S, Mammen JM, Padhye SB, Weir SJ, Jensen RA, Sitta Sittampalam G, and Anant S. Metastatic tumor-in-A-Dish, a novel multicellular organoid to study lung colonization and predict therapeutic response. Cancer Res 79:1681–1695, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Read GH, Miura N, Carter JL, Kines KT, Yamamoto K, Devasahayam N, Cheng JY, Camphausen KA, Krishna MC, and Kesarwala AH. Threedimensional alginate hydrogels for radiobiological and metabolic studies of cancer cells. Colloids Surf. B 171:197–204, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rianna C, and Radmacher M. Influence of microenvironment topography and stiffness on the mechanics and motility of normal and cancer renal cells. Nanoscale 9:11222–11230, 2017. [DOI] [PubMed] [Google Scholar]
  • 107.Rianna C, and Radmacher M. Comparison of viscoelastic properties of cancer and normal thyroid cells on different stiffness substrates. Eur. Biophys. J 46:309–324, 2017. [DOI] [PubMed] [Google Scholar]
  • 108.Rice AJ, Cortes E, Lachowski D, Cheung BCH, Karim SA, Morton JP, and del Río Hernández A. Matrix stiffness induces epithelial–mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis 6:e352–e352, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Romero-López M, Trinh AL, Sobrino A, Hatch MMS, Keating MT, Fimbres C, Lewis DE, Gershon PD, Botvinick EL, Digman M, Lowengrub JS, and Hughes CCW. Recapitulating the human tumor microenvironment: colon tumor-derived extracellular matrix promotes angiogenesis and tumor cell growth. Biomaterials 116:118–129, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Rossi G, Manfrin A, and Lutolf MP. Progress and potential in organoid research. Nat. Rev. Genet 19:671–687, 2018. [DOI] [PubMed] [Google Scholar]
  • 111.Sachs N, and Clevers H. Organoid cultures for the analysis of cancer phenotypes. Curr. Opin. Genet. Dev 24:68–73, 2014. [DOI] [PubMed] [Google Scholar]
  • 112.Sadtler K, Sommerfeld SD, Wolf MT, Wang X, Majumdar S, Chung L, Kelkar DS, Pandey A, and Elisseeff JH. Proteomic composition and immunomodulatory properties of urinary bladder matrix scaffolds in homeostasis and injury. Semin. Immunol 29:14–23, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Saito Y, Muramatsu T, Kanai Y, Ojima H, Sukeda A, Hiraoka N, Arai E, Sugiyama Y, Matsuzaki J, Uchida R, Yoshikawa N, Furukawa R, and Saito H. Establishment of patient-derived organoids and drug screening for biliary tract carcinoma. Cell Rep 27:1265.e4–1276.e4, 2019. [DOI] [PubMed] [Google Scholar]
  • 114.Saldin LT, Cramer MC, Velankar SS, White LJ, and Badylak SF. Extracellular matrix hydrogels from decellularized tissues: structure and function. Acta Biomater 49:1–15, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sawicki LA, Kloxin AM, Ross KE, Cowart JE, Pradhan L, Wu CH, and Ovadia EM. Tunable synthetic extracellular matrices to investigate breast cancer response to biophysical and biochemical cues. APL Bioeng 3:016101, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Seo Y, Jung Y, and Kim SH. Decellularized heart ECM hydrogel using supercritical carbon dioxide for improved angiogenesis. Acta Biomater 67:270–281, 2018. [DOI] [PubMed] [Google Scholar]
  • 117.Smithmyer ME, Spohn JB, and Kloxin AM. Probing fibroblast activation in response to extracellular cues with whole protein- or peptide-functionalized step-growth hydrogels. ACS Biomater. Sci. Eng 4:3304–3316, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Song H, Yang P, Huang P, Zhang C, Kong D, and Wang W. Injectable polypeptide hydrogel-based co-delivery of vaccine and immune checkpoint inhibitors improves tumor immunotherapy. Theranostics 9:2299–2314, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sun H, Su J, Meng Q, Yin Q, Chen L, Gu W, Zhang P, Zhang Z, Yu H, Wang S, and Li Y. Cancer-cell-biomimetic nanoparticles for targeted therapy of homotypic tumors. Adv. Mater 28:9581–9588, 2016. [DOI] [PubMed] [Google Scholar]
  • 120.Sun L, Wang Y, Cen J, Ma X, Cui L, Qiu Z, Zhang Z, Li H, Yang R-Z, Wang C, Chen X, Wang L, Ye Y, Zhang H, Pan G, Kang J-S, Ji Y, Zheng Y-W, Zheng S, and Hui L. Modelling liver cancer initiation with organoids derived from directly reprogrammed human hepatocytes. Nat. Cell Biol 21:1015–1026, 2019. [DOI] [PubMed] [Google Scholar]
  • 121.Suresh S Biomechanics and biophysics of cancer cells. Acta Mater 55:3989–4014, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Swaminathan S, Hamid Q, Sun W, and Clyne AM. Bioprinting of 3D breast epithelial spheroids for human cancer models. Biofabrication 11:025003, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Teng MWL, Ngiow SF, Ribas A, and Smyth MJ. Classifying cancers basedon T-cell infiltration and PD-L1. Cancer Res 75:2139–2145, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Theocharis AD, Skandalis SS, Gialeli C, and Karamanos NK. Extracellular matrix structure. Adv. Drug Deliv. Rev 97:4–27, 2016. [DOI] [PubMed] [Google Scholar]
  • 125.Tian B, Luo Q, Ju Y, and Song G. A soft matrix enhances the cancer stem cell phenotype of HCC cells. Int. J. Mol. Sci 20:2831, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tian H, Luo Z, Liu L, Zheng M, Chen Z, Ma A, Liang R, Han Z, Lu C, and Cai L. Cancer cell membrane-biomimetic oxygen nanocarrier for breaking hypoxia-induced chemoresistance. Adv. Funct. Mater 27:1703197, 2017. [Google Scholar]
  • 127.Tian YF, Ahn H, Schneider RS, Yang SN, Roman-Gonzalez L, Melnick AM, Cerchietti L, and Singh A. Integrin-specific hydrogels as adaptable tumor organoids for malignant B and T cells. Biomaterials 73:110–119, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Tlsty TD, and Coussens LM. Tumor stroma and regulation of cancer development. Annu. Rev. Pathol. Mech. Dis 1:119–150, 2006. [DOI] [PubMed] [Google Scholar]
  • 129.Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, Dwinell MB, Hunt B, Evans DB, Gershan J, and James MA. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer 18:335, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Tuveson D, and Clevers H. Cancer modeling meets human organoid technology. Science 364:952–955, 2019. [DOI] [PubMed] [Google Scholar]
  • 131.Walker C, Mojares E, and Del Río Hernández A. Role of extracellular matrix in development and cancer progression. Int. J. Mol. Sci 19:3028, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wang X, Zhang X, Dai X, Wang X, Li X, Diao J, and Xu T. Tumor-like lung cancer model based on 3D bio-printing. 3 Biotech 8:501, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wei SC, and Yang J. Forcing through tumor metastasis: the interplay between tissue rigidity and epithelial-mesenchymal transition. Trends Cell Biol 26:111–120, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Wippold FJ, and Perry A. Neuropathology for the neuroradiologist: rosettes and pseudorosettes. Am. J. Neuroradiol 27:488–492, 2006. [PMC free article] [PubMed] [Google Scholar]
  • 135.Wisdom KM, Adebowale K, Chang J, Lee JY, Nam S, Desai R, Rossen NS, Rafat M, West RB, Hodgson L, and Chaudhuri O. Matrix mechanical plasticity regulates cancer cell migration through confining microenvironments. Nat. Commun 9:4144, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wolf MT, Daly KA, Brennan-Pierce EP, Johnson SA, Carruthers CA, D’Amore A, Nagarkar SP, Velankar SS, and Badylak SF. A hydrogel derived from decellularized dermal extracellular matrix. Biomaterials 33:7028–7038, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wolf MT, Ganguly S, Wang TL, Anderson CW, Sadtler K, Narain R, Cherry C, Parrillo AJ, Park BV, Wang G, Pan F, Sukumar S, Pardoll DM, and Elisseeff JH. A biologic scaffold-associated type 2 immune microenvironment inhibits tumor formation and synergizes with checkpoint immunotherapy. Sci. Transl. Med 11:eaat7973, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Xu K, Ganapathy K, Andl T, Wang Z, Copland JA, Chakrabarti R, and Florczyk SJ. 3D porous chitosan-alginate scaffold stiffness promotes differential responses in prostate cancer cell lines. Biomaterials 217:119311, 2019. [DOI] [PubMed] [Google Scholar]
  • 139.Xu K, Lee F, Gao SJ, Chung JE, Yano H, and Kurisawa M. Injectable hyaluronic acid-tyramine hydrogels incorporating interferon-α2a for liver cancer therapy. J. Control. Release 166:203–210, 2013. [DOI] [PubMed] [Google Scholar]
  • 140.Xuan W, Qu Q, Zheng B, Xiong S, and Fan G-H. The chemotaxis of M1 and M2 macrophages is regulated by different chemokines. J. Leukoc. Biol 97:61–69, 2015. [DOI] [PubMed] [Google Scholar]
  • 141.Yom-Tov O, Seliktar D, and Bianco-Peled H. PEG-Thiol based hydrogels with controllable properties. Eur. Polym. J 74:1–12, 2016. [Google Scholar]
  • 142.Yu S, Wang C, Yu J, Wang J, Lu Y, Zhang Y, Zhang X, Hu Q, Sun W, He C, Chen X, and Gu Z. Injectable bioresponsive gel depot for enhanced immune checkpoint blockade. Adv. Mater 30:1801527, 2018. [DOI] [PubMed] [Google Scholar]
  • 143.Yu W, He X, Yang Z, Yang X, Xiao W, Liu R, Xie R, Qin L, and Gao H. Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis. Biomaterials 217:119309, 2019. [DOI] [PubMed] [Google Scholar]
  • 144.Yue B Biology of the extracellular matrix: an overview. J. Glaucoma 23:S20–S23, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zamani E, Shokrzadeh M, Fallah M, and Shaki F. A review of acrylamide toxicity and its mechanism. Pharm. Biomed. Res 3:1–7, 2017. [Google Scholar]
  • 146.Zhang Z, Qian H, Yang M, Li R, Hu J, Li L, Yu L, Liu B, and Qian X. Gambogic acid-loaded biomimetic nanoparticles in colorectal cancer treatment. Int. J. Nanomed 12:1593–1605, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Zhu Y, Hideyoshi S, Jiang H, Matsumura Y, Dziki JL, LoPresti ST, Huleihel L, Faria GNF, Fuhrman LC, Lodono R, Badylak SF, and Wagner WR. Injectable, porous, biohybrid hydrogels incorporating decellularized tissue components for soft tissue applications. Acta Biomater 73:112–126, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES