Abstract
Cancer research has considerably progressed with the improvement of in vitro study models, helping to understand the key role of the tumor microenvironment in cancer development and progression. Over the last few years, complex 3D human cell culture systems have gained much popularity over in vivo models, as they accurately mimic the tumor microenvironment and allow high-throughput drug screening. Of particular interest, in vitrohuman 3D tissue constructs, produced by the self-assembly method of tissue engineering, have been successfully used to model the tumor microenvironment and now represent a very promising approach to further develop diverse cancer models. In this review, we describe the importance of the tumor microenvironment and present the existing in vitro cancer models generated through the self-assembly method of tissue engineering. Lastly, we highlight the relevance of this approach to mimic various and complex tumors, including basal cell carcinoma, cutaneous neurofibroma, skin melanoma, bladder cancer, and uveal melanoma.
1. Introduction
Over the past years, various approaches have been employed to study cancer initiation, growth, and migration. Animal models have been instrumental in providing insight into the molecular mechanisms of tumor growth/proliferation and metastatic processes. However, studying cancer in vivo remains a considerable challenge nowadays, due to practical and ethical concerns, as well as to limitations in reliable predictions of human clinical trial outcomes [1]. Although current in vitro two-dimensional (2D) monolayer cell culture and conventional tridimensional (3D) cell culture systems have also led to significant advances in our understanding of tumor biology and the role of tumor microenvironment (TME), there are still several unmet needs to better model cancer invasion. The local TME is known to play a significant role in cancer progression and metastasis, where tumor cells can respond and adapt to a plethora of biochemical/biophysical signals from surrounding stromal cells and extracellular matrix (ECM) components [2]. In this regard, advances in tumor cell biology, 3D cell culture, and tissue engineering have enabled the rapid development of comprehensive in vitro tumor models with increased complexity, through the incorporation of multiple cell types. Furthermore, state-of-the-art tissue engineering technologies that incorporate endogenous patient-derived ECM proteins have emerged as unique alternatives to 3D bulk hydrogel and animal models to gain insights into the biological aspects of cancer development, which has not been fully possible using conventional culture systems. Other innovations including the incorporation of capillaries and ready-to-seed spheroids, grown under hypoxic or normoxic conditions, have led to precision medicine in the development of advanced tissue-engineered in vitro tumor models for patient-specific therapies, clinical management, and assessment of metastatic potential.
Among several protocols allowing the production of advanced exogenous material-free and patient-derived cancer models, the self-assembly method of tissue engineering distinguishes itself by the self-production and assembly of cell-specific endogenous ECM components. Indeed, it has been reported that ascorbate (vitamin C) can help mesenchymal cells produce their own ECM, to form a tissue highly similar to what is found in vivo [3]. Despite significant progresses over the last few years, there are still numerous challenges to create better models for various forms of primary and metastatic cancers, incorporating multicellular cultures and diverse cellular microenvironments capable of modulating ECM composition, cellular crosstalk, and distribution of soluble factors. In this review, we provide an overview of 3D cell culture models currently being employed with a particular focus on the stromal self-assembly method of tissue engineering, allowing the production of patient-derived organ- and human-specific models, for the study of diverse cancers.
2. Stroma and Tumor Microenvironment
2.1. Stromal Components
The stroma has long been viewed as a relatively inert structural support for organs. It is usually composed of connective tissue, the most abundant type of tissue in the body. It connects and supports other tissues and also plays a crucial role in organ development, homeostasis maintenance, and pathologies [4]. The connective tissue consists of cells, mainly fibroblasts, and ECM proteins. The specific ECM protein composition determines, in turn, the biochemical and biophysical properties influencing cell growth, differentiation, migration, and apoptosis [5–7].
The knowledge of the precise ECM composition of organ-specific human connective tissues is critical to better understand their structure-function relationship in healthy and diseased conditions. As evidenced for the skin dermis, the stroma can at least be divided into two distinct compartments: (1) the upper papillary dermis, a thin loosely arranged connective tissue, and (2) a deeper reticular dermis, consisting of a thick and dense irregular matrix [8]. These two distinct compartments have different biological roles but are also populated by distinct subsets of fibroblasts [9]. To accurately investigate pathological mechanisms, such as those occurring during cancer initiation and progression, generating innovative models including human organ-specific stroma has become essential. Among the cells inhabiting this rich environment, fibroblast is among the most abundant cell types present in the stroma, and is mostly responsible for the secretion and organization of the ECM.
Under stress conditions, fibroblasts adapt to their environment and have the ability to send local signals, to synthesize and reorganize the ECM of the skin and other organs [10, 11]. Fibroblast plasticity may be different depending on body parts, through a poorly understood mechanism involving the resident ECM framework and the microenvironment [12]. In certain pathological contexts, fibroblasts are capable of altering their cellular profile to become myofibroblasts, therefore producing massive amounts of ECM and contributing to organ dysfunction [13].
Many other cell types also live or transit through the stroma, including vascular endothelial cells, pericytes, adipocytes, and bone marrow stromal cells. Specialized cell types include nervous and immune cells. In addition to their classical roles, these cells can be unsuspected players in immune control [14] and wound healing [15]. Resident macrophages are especially sensitive to the matrix environment [16]. These cells could modify, directly or indirectly, through epithelial or endothelial cells, the secretion and organization of the ECM [17]. Some aspects of T cell memory function are now also associated with nonhematopoietic cells from the stroma [18].
2.2. Tumor Microenvironment
A plethora of different cell types located in the TME has been the focus of cancer research, including fibroblasts, myofibroblasts, endothelial cells, pericytes, macrophages, dendritic cells, and other immune cells. The nomenclature used to describe fibroblastic populations and fibroblast-like cells remains confusing because it widely varies in the literature. Most frequent terms include cancer-associated fibroblasts (CAFs), carcinoma-associated fibroblasts (also abbreviated as CAFs), tumor-associated fibroblasts, and tumor/cancer-associated stromal cells. A growing piece of evidence now shows that these cells are recruited from the stroma by cancer cells to promote ECM remodeling, neoangiogenesis, proliferation, invasion, migration, and metastasis, and mediate drug resistance mechanisms through the secretion of various growth factors, chemokines, and cytokines [19, 20]. Among these reactive stromal cells, CAFs are the most described and mainly derived from healthy fibroblasts, although they could also originate from mesenchymal stem/stromal cells (MSCs), adipocytes, endothelial cells, epithelial cells, or stellate cells [21]. They also share many similarities with wound healing-related myofibroblasts and differentiate when exposed to molecules such as transforming growth factor-beta, fibroblast growth factor 2, and platelet-derived growth factor [22]. CAFs are in constant interaction with cancer cells and have been shown to play a dual role in cancer progression, either promoting or suppressing it [23]. During tumor progression, CAFs enhance tumor growth through a variety of mechanisms, including ECM remodeling, and promote sustained inflammation via the increase of inflammatory cytokines, neoangiogenesis, and immune cell recruitment [24]. CAFs are now considered in many studies primary targets to limit cancer cell spreading [25].
Many cancers emerge from nonstromal tissues and migrate to the stroma, crossing the basal lamina in the case of epithelial cancers, or through intra- and extravasation processes in the case of circulating tumor cells [26]. The basal lamina is also present to delineate the endothelium, which separates two inner compartments, blood and connective tissues. It is composed of laminins [27], collagens (especially types IV and VII), and other molecules such as agrin, perlecan, and nidogens [28]. It is not only the physical support for epithelia and endothelia but also an important source of molecular cues that regulate cell interactions. Laminins are especially important because they anchor the cells to the basal lamina, through the cytoskeleton of epithelial cells or independently. They play a significant role as an interface between the epithelium and the stroma [28]. The presence of basal lamina is essential for the proper differentiation of epithelial cells, as evidenced by tissue engineering experiments where the inability of cells to deposit and assemble an organized basal lamina led to epithelial differentiation failures [29].
Other major constituents of the TME are ECM proteins. In healthy tissues, ECM is a dynamic microenvironment submitted to remodeling and degradation [30]. Deposited proteins include collagens, fibronectin [31], proteoglycans [32, 33], elastic fibres, and many others. They are mainly degraded by various enzymes including the matrix metalloproteinases (MMPs) [30]. In cancers, ECM is significantly modified to create an environment with different biophysical and mechanobiological features [34–36]. Cancer stroma is stiffer [37, 38], similar to what could be found in wound and fibrosis [39].It is also more prone to cell migration, due to a reduction of cell adhesion and epithelial-mesenchymal transition (EMT) [40].
2.3. Oxygen Tension in the Tumor Microenvironment
Each tissue is characterized by a specific oxygen partial pressure (pO2) in physiological conditions, namely, the in situ normoxia. This value varies from one tissue to another, as well as within the tissue itself, depending on the vasculature and the metabolic activity [41]. Tissue hypoxia is defined as an inadequate supply of oxygen that compromises biological functions. Since the uncontrolled proliferation of cancer cells causes tumors to outgrow their blood supply, most solid tumors have lower median pO2 than their tissue of origin [42]. Tumors are considered hypoxic when the pO2 falls below a critical value (8-10 mmHg) leading to a progressive decrease of the oxygen consumption rate or the ATP production rate in the tissue [43]. Adaptive responses to reduced oxygen availability are mediated by the hypoxia-inducible factors (HIFs), which are transcription factors active and stable under hypoxic conditions [44, 45]. HIFs function as master switches to induce the expression of several target genes involved in angiogenesis, cell survival, energy metabolism, differentiation, and invasion.
Although oxygen is supplied to the tissues and cells at reported concentrations of around 15-70 mmHg (2-9% O2) [46, 47], traditional tissue engineering practices rather use a pO2 that corresponds to the atmospheric level (160 mmHg or 21% O2). However, a physiologically relevant “low” pO2 could be beneficial for applications in tissue engineering [41]. For example, ECM synthesis and deposition by human dermal fibroblasts was shown to be optimal at a pO2 of 15 mmHg compared to cells exposed to a pO2 of 4 or 160 mmHg [48]. A low pO2 (15-40 mmHg) allowed the production of human vascularized cell sheets using the stromal vascular fraction of adipose tissue or MSCs without adding extrinsic growth factors [49, 50], as well as human tissue-engineered cartilaginous tissues using synovial MSCs without the use of an exogenous scaffold [51]. Also, stiffer intervertebral discs with increased glycosaminoglycan and collagen contents were generated when human MSCs were grown under hypoxic conditions (40 mmHg) during their 2D expansion and subsequent 3D culture [52].
Microenvironmental conditions such as oxygen tension and tissue dimensionality are critical determinants of tumor angiogenesis, fibrosis, and heterogeneity. Interestingly, many angiogenic factors and proteases that degrade or remodel the ECM, such as MMPs and lysyl oxidase, are among the genes comprising hypoxia-responsive elements in their promoter [53, 54]. Pathologically relevant 3D tumor invasion models, such as tumor spheroids with a hypoxic core embedded in self-assembled stromas, are thus required to better mimic the complex biophysical features of the TME and the tumor heterogeneity.
3. 3D Cell Culture and Spheroids as Cancer Models
2D cell culture has brought relevant information on cancer cell behavior and is still widely used for the prediction of drug responses, despite obvious limitations. Indeed, this simple and low-cost in vitro culture system has failed to properly model essential characteristics of tumors and TME such as the complex process of tumor invasion [55, 56]. 3D cell culture is an in vitro system that allows the growth and interaction of different cell types in a 3D scaffolding using either native ECM or biomaterials. Compared to conventional 2D cell cultures grown on plastic substrates, 3D systems better mimic the in vivo conditions in terms of cell-cell and cell-matrix interactions, physicochemical properties, and mechanical stresses [57–59]. 2D systems only offer weak interactions between cells and their microenvironment due to the inert nature of plastic. Moreover, cells grown as monolayers stay in a planar state, adhere and proliferate horizontally, and keep an apical-basal polarity [60, 61], which impedes or alters several biological processes, including differentiation and stemness. For example, few studies have demonstrated that freshly isolated cells can generate dedifferentiated cell populations when grown in 2D systems, while they can recover their native phenotype and primary functions when cultured in 3D environments [62, 63].
Interestingly, 3D culture systems can elicit a more physiological orientation of extracellular receptors, such as integrins and collagen receptors, for better interactions with neighbouring cells and ECM, compared to 2D systems [64]. They can also promote essential biological processes such as cell differentiation, migration, adhesion, proliferation, and morphogenesis [65]. While cells cultured in monolayers proliferate until they reach contact inhibition, cells in 3D develop in every direction and thus reach higher proliferation rates until homeostasis , especially when cocultured with stromal fibroblasts [66–68]. Consequently, 3D culture systems have progressively emerged as promising tools for the study of complex cellular interactions and now represent unavoidable models for clinical translation.
Due to the strong interaction between cancer cells and TME, 3D systems probably represent the best in vitro models for the study of cancers and the screening of innovative anticancer drugs. Therefore, 3D cultures may also offer a relevant platform to recreate and study the migratory behavior and invasion profile of cancer cells [69, 70], notably due to the expression of integrins and MMPs that resemble the native tissue [71]. Gene expression, mRNA splicing, and the biochemistry of cancer cells grown in 3D cultures are also similar to in vivo conditions [72, 73]. For example, the 3D culture of melanoma cells highly modulates gene expression with the notable upregulation of few CXCL genes, which have been linked to the progression and metastatic process of this cancer [72]. Moreover, 3D environments give rise to cancer cell behaviors that closely mimic those found in vivo [74–76]. Eventually, 3D cancer cell systems represent realistic models to monitor the effects of drug dosing and distribution [77].
Over the years, numerous 3D models have been developed to recreate the TME and study the cellular responses to drug treatments, including microfluidics, transwell-based assays, organoids, biogels, organs-on-a-chip, perfused cultures, bioreactors, and 3D bioprinting [78, 79]. These 3D models allow various degrees of complexity and may provide precious information on tumor formation, progression, and invasion, as reviewed elsewhere [80, 81]. More recently, 3D tumor spheroids have been extensively used in cancer research, especially in breast, lung, prostate, and colorectal cancer studies [81, 82]. These adaptative multicellular complex systems [83], composed of proliferative and quiescent cells, can reproduce highly interactive environments and closely mimic native tumor behaviors with similar histopathological features [82]. Depending on their size, they can carry a hypoxic or necrotic core, which is usually found in tumors [84]. Spheroidal structures also generate local nutrient, metabolite, and oxygen gradients, which help quantify the penetration of drugs [85].
3D tumor spheroids can be produced using various methods including the hanging drop technique and the bioreactor rotative system. The hanging drop technique can be used manually with an inverted dish or with specialized plates called ultralow attachment plates [82]. This method generates spheroids with rather similar sizes regardless of the cell line. However, this technique is time consuming, in spite of recent works speeding up the fabrication process [86]. In contrast, rotative systems allow the production of massive amounts of spheroids but are expensive and create heterogeneous populations of spheroids [82]. More complex techniques are available but require expensive equipment and trained staff. Among them, the production of spheroids within 3D-printed scaffolds has gained popularity, since it provides a complex 3D support. However, this approach sometimes integrates exogenous materials and causes shear stress on cells [87, 88]. In summary, the 3D tumor spheroid model is suitable for certain study purposes but can be limited by its production method, the capacity of aggregation, and maximal viable time in vitro. In the following sections, we will, therefore, focus on the alternative use of 3D tumor spheroids integrated into tissue-engineered cancer models.
4. The Self-Assembly Approach of Tissue Engineering
When cultured under optimal in vitro conditions, isolated primary stromal cells can constitutively secrete their own ECM proteins to promote the establishment of the cellular microenvironment. In vivo, stromal cells produce most of the ECM molecules that provide structural and biochemical support for surrounding cells [89]. We have taken advantage of the capacity of primary stromal cells to promote their microenvironment in vitro using the self-assembly method (Figure 1) [90] that relies on the long-term supplementation of serum and ascorbic acid in the culture medium. Ascorbic acid, a cofactor of the enzyme prolyl-hydroxylase, facilitates the hydroxylation of proline that is crucial to stabilize the collagen fibrils and their deposition. Secreted ECM components accumulate and assemble within the culture, resulting in a tissue sheet that is sturdy enough to be manipulated with forceps. These cell sheets can be superimposed to generate thicker and planar mesenchymal tissue that can be cultured for several weeks. The self-assembly approach allows the reconstruction of tissues that are histologically and physiologically similar to their in vivo counterparts [91]. This method has been successfully used to reconstruct numerous human tissues such as blood vessels [91, 92], bilayered skin [3], bladder [93], adipose tissue [94], heart valve [95], ureter [96], and cornea [97]. It also has been demonstrated that this technique allows to preserve subtle differences in native cells, such as receptor expression patterns, and reproduce pathologies, including psoriasis [98], fibrosis [99], amyotrophic lateral sclerosis [100], inflammation [101], and cancer [102, 103].
4.1. Cell Culture and Tissue Production
Cells can be either obtained from nonprofit organizations or isolated from fresh human tissues after obtaining the patient's consent. These tissues are usually collected following elective surgeries (normal skin, scars) or biopsies performed by a specialist. In this case, the biopsy is cleaned with sterile saline containing antibiotics, and then mechanically and/or enzymatically dissociated to digest the ECM and the basal lamina [104–107]. Of particular interest, it is possible to isolate several cell types from a single biopsy to create patient-derived tissue-engineered constructs to study disease-specific microenvironments. Different cell types can be cultured alone or in combination with one another, either isolated from healthy donors or from different patients to decipher the role of a specific cell type in the disease pathological process. To obtain cell and matrix-rich sheets that can be stacked upon one another and easily handled, stromal cells are first seeded on a tissue plate in the presence of a peripheral anchorage device (Figure 1(a)). This support limits the contraction of the tissue and facilitates the manipulation of the resulting cell sheet. Stromal cells are classically cultured in Dulbecco's Modified Eagle's medium containing 50 μg/ml ascorbate, 10% bovine serum, and antibiotics. After 14 to 28 days, several sheets are stacked to form a thicker stromal compartment. Then, and if needed, for skin or bladder reconstruction, for example, epithelial cells can be seeded on the surface of the reconstructed stroma. Afterward, such bilayered tissue constructs are cultured in an epithelial medium containing 50 μg/ml ascorbate, bovine serum, and additives for 4 to 7 days [3]. The construct is next transferred on supportive components to maintain the tissue at the air-liquid interface and further cultured for 10 to 21 days to allow a proper differentiation of the epithelium. Cancer cells or ready-to-seed 3D spheroids, prepared in parallel, can also be added into the tissue-engineered constructs during the reconstruction process (Figure 1(b)) [102, 103].
4.2. Addition of Vascular and Lymphatic Microvascular Networks in Self-Assembled Tissues
Initially, the rationale of adding a microvascular network to self-assembled tissues was to accelerate their perfusion by the host vasculature upon grafting. The underlying hypothesis was that grafted tissues that are more rapidly vascularized will not suffer from alterations due to a lack of oxygen and nutrients. To generate tissue-engineered constructs with microvascular networks, the first step is to have access to endothelial cells, which can be isolated from human umbilical vein, neonatal foreskin, and adult skin (reviewed in [108]). Then, endothelial cells must be cocultured with stromal cells or added to the cell sheets during the maturation phase (Figure 1(c)). When seeded on top of stromal cell sheets, they attach randomly to the surface and quickly form clusters within 48 hours. Tubular-like structures with lumens then emerge, forming spontaneous microvascular networks that persist over several weeks in vitro [109, 110]. These microvascular networks are functional when grafted. Indeed, prevascularized self-assembled tissue-engineered skin, adipose tissue, blood vessels, and urethra were shown to be perfused within 2 to 4 days following in vivo tissue implantation [96, 101, 111, 112]. In contrast, the detection of red blood cells was delayed within the grafts of control tissues without preestablished capillaries (e.g., between 7 and 14 days after grafting). Hence, the presence of a microvascular network accelerates the integration of the grafted tissues.
Of particular interest, prevascularizing reconstructed tissues allows to better understand normal and pathological processes of vascular and lymphatic vessel formation and remodeling, including in cancer. In self-assembled skin substitutes, the epidermis was shown to influence the diameter of dermal capillaries through a process involving the vascular endothelial growth factor [110]. The recruitment, from fibroblasts, of pericyte-like cells expressing α-smooth muscle actin or chondroitin sulfate proteoglycan, was also observed [109]. Since pericytes are essential components of the blood microvasculature, the self-assembly method of tissue engineering becomes a powerful approach to study angiogenesis and endothelial cell/pericyte interactions with the stroma in vitro. In the field of cancer research, the intima of tissue-engineered blood vessels was shown to react to the presence of the interleukin-1β inflammatory cytokine by expressing E-selectin, a binding molecule for circulating cancer cells [109, 113, 114]. Hence, the prevascularization of reconstructed tissues constitutes an interesting tool to investigate normal and tumoral mechanisms.
In many cancers, a common route for metastasis is the invasion of lymph nodes through the lymphatic vessels. It has been shown that lymphatic endothelial cells seeded within tissue-engineered constructs assembled into capillaries exhibiting nearly all of the molecular and ultrastructural features of native human lymphatic microvasculature, including branching in 3D, wide lumen, blind ends, overlapping borders, adherens junctions, anchoring filaments, lack of mural cells, and poorly developed basement membrane [115]. Lymphatics within tissue-engineered constructs also form more tubules in response to lymphangiogenic stimuli [116]. Overall, these models hold promises to decipher the pathophysiology of cancer because the presence of a microvascular network contributes to the process of metastatic progression [117].
5. 3D Cancer Modeling Using the Self-Assembly Approach
Cancer modeling is useful for basic and applied research, giving insights into the pathophysiology of tumors and creating novel tools for drug discoveries. As mentioned above, 3D cancer models can be used to study the TME, while 2D culture systems cannot. The understanding of the role of specific fibroblast subtypes, as well as ECM organization, fibre size, orientation, and stiffness, is a crucial aspect in cancer research which has led to the development of novel 3D culture systems using exogenous scaffolds [89]. The heterogeneity and plasticity of stromal cells across the body indeed result in the formation of matrix microenvironments with distinct composition and organization, as illustrated in the papillar and reticular dermis of the skin [8]. These distinctive matrix microenvironments are likely to influence cancer behavior, development, and spreading [118, 119], as shown with the variety of metastatic mechanisms described in prostate [120], colorectal [121], breast [122], bone [123, 124], and skin [125] tissues.
With the development of self-assembly approaches, it has become possible to recreate specific human matrix microenvironments in vitro. Self-assembled tissues indeed contain organ-specific stromal cells that provide the proper ECM composition and organization to induce physiological processes, such as cornea [126] and bladder [127] epithelial cell differentiation. When fabricated from pathological cells, self-assembled tissues display abnormal ECM networks resembling native pathological matrices, as shown in reconstructed models of hypertrophic scars [98], scleroderma [128], and psoriasis [98]. In this section, we will show that self-assembled tissues can also mimic the TME of native human skin, bladder, and eye cancers, therefore providing highly valuable study models for cancer research and precision medicine.
5.1. Basal Cell Carcinoma
Basal cell carcinoma (BCC) is one of the most prevalent cancers in the world, accounting for 60-80% of all nonmelanoma skin cancers [129]. Most frequent etiologies in patients with BCC include genetic predispositions, chronic ultraviolet (UV) exposure, and nonhealing wounds such as ulcers and severe burns [130]. Usually, BCC manifests in body areas covering the head and neck (80% of cases), the trunk (15% of cases), and the arms and legs. More rarely, it also arises from the armpits, breasts, perianal areas, palms, and soles [131]. Clinically, BCCs are grouped into indolent and aggressive growth forms depending on the patient genetic background. The indolent variants, which are the most common subtypes , include nodular and superficial BCCs, while the aggressive variants comprise micronodular and morpheaform subtypes. Although BCCs are rarely associated with metastases and mortality, they can spread locally and damage the cutaneous tissue, leading to loss of function and disfiguring outcomes [132]. Over the last 50 years, the incidence of BCC has been rising in Europe [133, 134], Asia [135, 136], and America [137, 138], reaching up to 1.5% of the male population in the US. Unsurprisingly, the management of BCC has thus become a significant burden for most healthcare systems. In the US, for example, radiation therapy and surgery treatments can cost over $3,000 per patient [139].
In the mid-19th century, seminal studies, driven by Robert Gorlin, in patients with basal nevus syndrome were the first to suggest a genetic etiology for BCC [140]. Since then, genetic knockout mice havebecome the primary tool to study the disease, based on the fact that BCC patients suffer from a mutation in the Patched 1 (PTCH1) gene, dysregulating the Sonic Hedgehog (SHH) pathway [141, 142]. The first transgenic mice were developed to overexpress the SHH protein [143], or a mutant variant of the downstream protein SMO [144]. However, these mutations were lethal and fostered the development of alternative strategies. The successful overexpression of SHH signaling transcription factors, such as Gli-1 [145] and Gli-2 [146], and the development of UV radiation-sensitive models using PTCH+/- transgenic mice [147] constituted robust in vivo models to study BCC. However, considering the distant relationship between rodents and humans, it became more and more questionable whether these models were truly reflecting the effects observed in humans. A few years ago, the SMO antagonist vismodegib, approved by the Food and Drug Administration for the treatment of aggressive BCC, was indeed discontinued because of its cumulative toxicity in patients [148]. This example shows that the use of study models that more closely mimic human physiology is an absolute prerequisite to improve the screening of new BCC therapies.
Over the past few years, discoveries in cancer research have pointed out several factors implicated in the BCC pathophysiology, including mutations in the tumor-suppressing gene p53 [149], activation of the Wnt/β-catenin pathway [150], and signaling through the epidermal growth factor receptor [151]. The TME has also been shown to influence BCC growth and progression, through the release of oncogenic proteins such as Gremlin-1 [152] and Galectin-1 [153]. Despite the need for in vitro models that recapitulate the human TME and bypass the use of mouse models, there still is a lack of accurate human BCC 3D models, since most of the current research uses animal tissue explants [154] or 2D cell culture.
In this context, the stromal self-assembly method of tissue engineering stands as a promising approach to faithfully recreate the human BCC pathophysiology. Using this method, successful reconstruction of pathological BCC skin constructs has been achieved in 56 days using cells derived from BCC patients (Figure 2(a)). Briefly, fibroblasts were isolated from BCC patients and cultivated with ascorbate until they formed cohesive stromal sheets. These sheets were then stacked to one another, forming a dermal construct, on top of which normal and BCC keratinocytes were seeded and matured at the air-liquid interface. Using this model, BCC skin constructs displayed histological features of BCC cancers, including nests of basaloid cells , surrounded by a fibromyxoid stroma (Figures 2(b)–2(i)). Tumoral keratinocytes also showed abnormal proliferating and nondifferentiating phenotypes compared to the control, as depicted by the divergent expression patterns of K10 and K15 (Figures 2(c), 2(d), 2(g), and 2(h)). The aberrant expression of the basement membrane protein, type IV collagen, around the dermal nests of BCC skin constructs further confirmed the basaloid phenotype of pathological cells (Figures 2(e) and 2(i)).
Although requiring long production times and staff who are well trained in complex cell culture techniques, the stromal self-assembly method of tissue engineering represents a reliable and promising tool to produce human BCC tissues in vitro. Compared to other conventional in vitro models, the stromal self-assembly method allows the development of a complex 3D TME, essential to recapitulate the histology of human BCC in vitro. Additionally, it is neither limited by tissue donor availability nor ethical concerns, as it is often the case with human tissue explant culture models. It can also be used to investigate the role of various targets since cultured cells can undergo any modification before being incorporated into the 3D model.
5.2. Cutaneous Neurofibromas
Neurofibromatosis type 1 (NF1) is a common genetic disorder of deregulated cell growth, occurring in about 1 in 3,000 individuals, caused by germline mutations in the neurofibromin-encoding gene NF1 [155]. People with NF1 are at an increased risk of developing a variety of benign and malignant tumors. There are limited therapies and no cures for NF1. It causes brain tumors, as well as disfiguring skin lesions called cutaneous neurofibroma (cNF) that stigmatize the affected individuals. The morphogenesis of neurofibromas is poorly understood and their formation can highly vary between patients, with significant differences sometimes seen within the same family. The presence of cNFs is one of the main clinical features of patients affected by this disease. Neurofibromin is a tumor suppressor protein involved in the regulation of the RAS/MAPK signaling pathway [156–158]. Typically, cNFs are benign tumors developing from peripheral nerves that form rounded and pedunculated masses within the dermis [159–161]. They are principally composed of Schwann cells, fibroblasts, endothelial cells, and mast cells, all embedded in a dense collagen-rich ECM [162–164]. In addition to cNF, NF1 patients can develop plexiform neurofibromas (pNFs) that can undergo malignant transformation and form a malignant peripheral nerve sheath tumor (MPNST) [159].
The biallelic NF1 inactivation in Schwann cells is suggested to be the first step of cNF formation, but the exact biological mechanism remains poorly understood [165, 166]. Complete loss of NF1 function alone cannot explain tumor development, and several experimental studies suggest that other factors such as the NF1 haploinsufficient cellular type or the stromal microenvironment may also be involved. Clinical presentation of cNFs is highly heterogenous and considerably differs from patient to patient in terms of number, size, occurrence, and localization [167, 168]. In numerous cases, neurofibromas instigate significant disfigurement and discomfort, causing psychosocial problems among NF1 patients. Although significant progresses have been made in the understanding of NF1 biology, patients still face significant morbidity and decreased life span. The pathogenic mechanisms of cNF formation and progression are still largely unknown, and there is no specific treatment for this complication. Traditional 2D cell culture models, as well as NF1-derived animal models, failed to recapitulate this important aspect of the disease manifestations [169–171]. Kraniak et al. have developed 3D models where immortalized NF1-associated cells isolated from pNF formed spontaneously spherical aggregates when seeded in Matrigel [172]. They also tested different drugs and observed resistance to growth inhibition in cells cultured in 3D. Other groups have demonstrated that induced pluripotent stem cell- (iPSC-) derived NF1 Schwann cells formed 3D spheroid-like structures in culture and expressed specific pNF markers [173]. Indeed, these 3D models are particularly interesting and beneficial for the NF1 field, but they are not necessarily suitable for long-term studies. Also, the ECM scaffold used for 3D cell culture does not represent the TME usually found in NF1-associated neurofibromas.
It has been recently shown that patient-derived spheroids, composed of NF1+/- Schwann cells alone (Schwannoma) or cocultured with NF1+/- fibroblast cells, inoculated within tissue-engineered 3D skin substitutes, led to the formation of cNF with histopathological characteristics reminiscent of native tumors [174]. Thus, this innovative patient-derived 3D cNF model could allow to better study neurofibroma morphogenesis and stimulate research on this topic. The well-known hanging drop method was used to generate the abovementioned patient-derived spheroids. Three days before the superposition of 3D fibroblast cell sheets, well-formed spheroids were seeded on the top of the previously generated NF1-derived skin substitutes using the self-assembly method of tissue engineering (Figure 3). The full 3D cNF tissue-engineered model was obtained after 52 days of culture. Interestingly, spheroid composition and cellular concentrations can easily be modulated before the incorporation within the tissue-engineered dermis. Different ratios of each cell type can also be used, and different cell types normally found in neurofibromas, such as endothelial cells and mast cells, can be added within the 3D spheroids to recreate a vascularized TME (Figure 3(d)).
This model can be used to study the cell of origin leading to the formation of cNFs and clinical heterogeneity associated with different gene mutations. Finally, this 3D cNF model could have a considerable impact on the understanding of pathogenic mechanisms underlying NF1 and become anajustable platform to accelerate drug discovery and prevent the development of these tumors.
5.3. Skin Melanoma
Melanoma is the deadliest skin cancer. In 2015, its global incidence was estimated to 351,880 cases with an age-standardized rate of five cases per 100,000 persons [175]. According to the National Cancer Institute, there were about 96,480 new melanomas diagnosed in 2019 in the US alone, and 7,230 people are expected to die of this cancer [176, 177]. Cumulative sunlight exposure has been described as the major etiological factor in melanoma [178, 179], and DNA damages that are created by UV radiation are also prognostic for outcomes [180]. Currently, diagnosis at an early stage and surgical ablation of the tumor is the best avenue to maximize patient survival [177].
Skin melanoma originates from the malignant transformation of melanocytes, which are located in the basal lamina of the epidermis and hair follicles. From a histopathological perspective, tumor development in the primary melanotic lesion begins by two successive phases: a horizontal phase, which is characterized by uncontrolled cell growth, followed by a vertical phase, in which EMT eventually occurs, promoting malignancy [181, 182]. Crosstalk between melanoma cells and the TME, including immune cells, further influence metastatic progression, which ultimately results in the invasion of capillaries and dissemination to distant sites.
To study and understand skin melanoma in vitro, it is important to recreate the native tissue. In recent years, there have been several attempts to include specific components of the melanoma TME to better study its pathophysiology [183, 187]. Indeed, such models can help elucidate invasive mechanisms of this cancer through its interaction with fibroblasts and keratinocytes [184, 185]. In an interesting report, Morales et al. assessed the sensitivity of melanoma cells to the B-Raf inhibitor vemurafenib in a 3D coculture system consisting of metastatic melanoma cells incorporated in a dermal fibroblast-derived matrix scaffold [186]. In the field of oncopharmacology and drug delivery, basic spheroid models have been used to study the potential of microparticles to deliver drugs [187]. Interestingly, spheroids can also be added to 3D skin models to create moles. Such 3D organotypic skin-bearing melanoma spheroids have been useful to investigate anticancer drugs in an environment that recreates a major part of the complexity of the tumor [186, 188].
The addition of a microvascular network to 3D models is necessary to recapitulate the full extent of the metastatic process. The presence of a microvascular system was shown to be required to trigger the vertical invasion of melanoma cells in vivo [117]. Because microvascular networks are not present in the majority of skin melanoma models developed to date, their influence has been investigated using a self-assembly approach. Gibot et al. have reported that an angiogenic effect is induced by the presence of melanoma cell lines seeded randomly in 3D skin models [189]. To further recreate the native heterogenous tumor microenvironment , melanoma cells seeded as tumor spheroids were also added to this model in addition to both vascular and lymphatic capillaries (Figure 4). This recent development allows for the evaluation of potential therapeutics and novel drugs in melanoma progression in a complex environment exempt from exogenous material. Of particular interest, Bourland et al. have investigated the impact of chronic treatment with vemurafenib in the model and observed a dose-dependent response on proliferation and apoptosis [102].
The protocol to investigate melanoma through this approach required 7 weeks. In the first week, cells were thawed and amplified. In the second week, fibroblasts were seeded on a paper anchor and cultured in the presence of ascorbate to produce macroscopic living cell sheets that could be manipulated with forceps. Melanoma spheroids were also produced in the second week, and endothelial cells, as well as keratinocytes, were thawed and amplified. On the 21st day, endothelial cells and melanoma spheroids were seeded onto the fibroblast cell sheets. On the 24th day, keratinocytes were seeded on a few number of fibroblast cell sheets. One week later, two cell sheets without keratinocytes were stacked under a cell sheet seeded with keratinocytes and placed at the air-liquid interface. The complete models were considered mature (stratified epidermis, incorporated tumor, pigmentation, and capillary networks) at day 38, and experimental treatments, such as the exposure to UV radiation, could be investigated. Of particular interest, numerous primary human or metastatic melanoma cell lines are commercially available and can be added as spheroids in the model. The metastatic potential of these cancerous cells can be assessed in tissue-engineered skins with or without the addition of oncogenic and/or prometastatic factors such as UV radiation.
To summarize, 3D skin models of melanoma have the potential to provide relevant information on melanoma progression, invasion, and response to therapeutics. These models may constitute interesting alternatives to animal studies by allowing them to investigate specific questions [183, 187]. Hence, it is reasonable to envision that 3D skin models will be increasingly used as research tools to decipher specific mechanisms related to metastatic progression and the development of therapeutics in melanoma.
5.4. Bladder Cancer
Bladder cancer is a recurrent and malignant disease that afflicts around 550,000 patients per year. It is the 10th most common cancer [190]. Bladder cancer affects more men than women with a sex ratio of 3 : 1 [191]. The main cause of bladder cancer is environmental or occupational exposure to carcinogens, especially tobacco [192]. It is characterized by a great level of recurrence and increased aggressiveness upon return.
The bladder consists of four different layers: from the outer to the lumen, (1) the adventice which consists of a connective tissue roughly similar to the adipose tissue, (2) the muscular layer called detrusor which has the function of expelling the urine outside the bladder during the muscle contraction micturition phase, (3) the submucosa layer called lamina propria which consists of a connective tissue joining the detrusor and the fourth layer, and (4) the urothelium. The urothelium is the bladder epithelium separated from the rest of the bladder by the basal lamina. It consists of 5 to 7 layers of pseudostratified bladder urothelial cells (BUCs); the basal layer, which contains the stem/progenitor cells and poorly differentiated BUCs; and the intermediate layers, 3 to 5 layers depending of the filling status of the bladder, which consists of elongated cells attached to the basal lamina, the most basal cells being less differentiated than the most apical ones. Finally, the superficial layer is found directly in contact with the urine, and consists of large, flat, frequently binucleated cells. These cells are called umbrella cells and cover several intermediate cells. They express and assemble uroplakins which form plaques of proteins to protect the epithelium from the urine, a toxic and potentially carcinogenic liquid waste. The urothelium is in constant interaction with the underlying lamina propria to maintain tissue homeostasis, adequate proliferation rate, and differentiation of BUCs [193–196].
To study and understand bladder cancer, it is important to work with a model exhibiting similar properties to the native tissue. Several assays were made for three-dimensional bladder cancer modeling, especially spheroid and organoid models [197–199]. Recent publications have highlighted the importance of the chosen protocol to produce spheroids [200, 201] and their usefulness compared to two-dimensional cell line cultures [202]. Also, the organoid model of bladder cancer allows demonstrating the involvement of the Wnt/β-catenin pathway in cancer progression [203]. Interestingly, long-term three-dimensional models from patient-derived primary cancer cells have been established as powerful models to potentially develop personalized treatment [204–206].
A self-assembly protocol has also been used to model bladder cancer. Originally, the goal of developing a 3D bladder model was to address a need to increase the bioavailability of tissues for bladder augmentation by generating tissue-engineered autologous substitutes. Since numerous reconstruction techniques use biomaterials or exogenous materials to obtain such models, cell differentiation and function may not be physiologically accurate, limiting their grafting potential [107].
The self-assembly technique was used to obtain a completely human 3D bladder model. Stromal cells were used to secrete and form the ECM, and urothelial cells were then seeded directly on the stroma [127]. Fibroblasts from a skin biopsy were generally used to produce these models. Results showed that nonbladder mesenchymal cells altered urothelium differentiation. The urothelial cells presented epithelial markers, such as cytokeratin 14, after they were cultured on the stromal compartment. Therefore, the use of organ-specific stromal cells was tested to improve and normalize the urothelium differentiation and eliminate this problem. The bladder mesenchymal cells, contrarily to dermal fibroblasts, promoted the normal differentiation of urothelial cells, eliminating the presence of epithelial markers. This reconstructed tissue was first produced in a porcine-derived model [127], followed by a human-derived model [103].
To study the development of bladder cancer cells in vitro, it is important to reproduce a valid tumor microenvironment. To achieve such a feat, all cells used in the self-assembly technique have to be isolated from a human bladder biopsy. Three sheets of fibroblasts and their ECM were created in separate culture dishes and superimposed to obtain the thickness needed for our 3D model. To promote ECM formation, fibroblasts were stimulated with ascorbate and serum over 21 days. After the stacking of the sheets, urothelial cells were cultured on the surface sheet. After 4 days of culture in medium, the tissue was raised to the air-liquid interface to mimic the conditions in which urothelial cells differentiate for 21 days. Finally, to assess cancer development, spheroids made from cancer cells were deposited on the newly differentiated urothelium (Figure 5(a)). It is important to note that for the cancer model to be relevant, a basement membrane must have appeared between the urothelium and the stroma. The basement membrane plays a great role in cancer progression and epithelial-mesenchymal transition. This detail is often neglected in invasive cancer studies [103].
The obtained model showed many similarities to a native human bladder mucosa. Type I collagen was detected in the stroma, along with CD31, indicating the potential presence of capillary-like networks. Laminin 5 was detected in both the reconstructed model and the native human bladder; it is an important marker showing the presence of a basement membrane. Cytokeratins were also detected in the urothelium of both native and reconstructed tissues [103]. The spheroids made from noninvasive cells did not penetrate beyond the urothelium, just as what would be observed in native tissues, whereas the muscle-invasive bladder cancer cells were able to penetrate the urothelium and completely disorganize the basement membrane. In both cases, the results were similar to what would be seen in vivo (Figure 5). It has been proven that it is possible to obtain a self-assembled tissue including the stroma and a differentiated epithelium, separated by a basal lamina. This model is a great tool for tumor progression studies and drug development.
5.5. Uveal Melanoma
Uveal melanoma (UM) is the most frequent primary intraocular tumor in adults and is drastically distinct from skin melanoma in terms of etiology, biology, and metastatic organotropism [207, 208]. It arises from the malignant transformation of ocular melanocytes, with a higher occurrence in the choroid coat [207, 209]. The most common age at diagnosis is 50-60 years, and the incidence is highest among people with lighter skin, fair hair, and blue eyes [210]. Familial cases and germline mutations are rare in UM [211, 212]. Key chromosomal abnormalities and driver mutations, as well as the DecisionDx®-UM gene expression profile test allow classifying UM patients at low or high risk of metastasis [213]. Indeed, half of the patients develops metastases mostly in the liver (so-called hepatic tropism). The ocular tumor is generally treated by radiation therapy or eye enucleation [214, 215]. However, metastatic disease is incurable. Contrary to skin melanoma, only a minority of patients benefited from immunotherapy [216, 217].
The self-assembly approach of tissue engineering will be of great interest to define the role of the microenvironment in UM, at both the primary and metastatic sites. A tissue-engineered choroid model was recently generated to address the need for 3D substitutes to study diseases affecting this ocular tissue such as age-related macular degeneration. This model includes choroidal fibroblasts with their endogenous ECM, as well as choroidal melanocytes [218], endothelial cells, and the retinal pigment epithelium, and successfully recaptures the biomechanical properties of the native tissue [219]. This innovative 3D system is superior to previously published in vitro models of the choroid, which used synthetic scaffolds or primate choroidal endothelial cells [220, 221]. UM-like spheroids were also seeded in this tissue-engineered choroid model to decipher the pathological contribution of the choroidal microenvironment in UM progression [222] and to eventually provide a valuable tool to improve the treatment of the primary tumor.
The development of new therapeutic strategies to cure the metastatic stage of UM will require a better understanding of mechanisms that allow the stromal liver cells, such as hepatic stellate cells (HSteCs), to create a permissive environment [223]. Once activated into a myofibroblastic phenotype, they can prime the premetastatic niche by remodeling the hepatic ECM as demonstrated in metastatic pancreatic and colorectal cancers [224, 225] and more recently in metastatic UM [226, 227]. Also, their fibrogenesis activity may be unfavorable to the efficacy of several anticancer treatments [228]. It is becoming clear that recreating the liver-specific human ECM, both composition and architecture, as well as hepatic cell-matrix/cell-cell interactions, will be difficult but essential for liver metastasis studies [229]. Human liver organoids were recently developed using iPSCs [230]. However, this approach is technically challenging and too expensive to be used as routine 3D models in both the academic and pharmaceutical industry. It is well recognized that synthetic materials and matrices formed from isolated biological materials cannot achieve the molecular complexity and organization of native tissue or tumor matrices. Since UM patients' histological samples correspond mostly to the macrometastatic disease, an in vitro model of metastatic UM will allow characterizing the ECM remodeling at both early and late metastatic stages, in addition to xenograft models [226]. The development of HSteC-derived matrices incorporating metastatic UM spheroids is underway to uncover how the desmoplastic reaction of stellate cells contributes to the notorious therapeutic resistance of UM [218, 231]. The strategy of combining the self-assembled cancer organotypic model and microfluidics to generate cancer-on-a-chip models [232] may eventually greatly contribute to predicting drug-induced responses in metastatic UM.
6. Perspectives and Future Directions
In this review, we have discussed current advances in 3D in vitro cell culture to study cancer. In particular, we described cancer models engineered to study a variety of tumor types, including those developing in the skin, bladder, and eye tissues. Other important cancer types for which solid 3D tumor models are being developed by various teams include prostate and breast cancers [20, 233–240]. Recent advances for these tumor types include the use of organoids and the incorporation of patient-derived samples (blood derivatives, cells, and/or ECM). The increased use of primary patient-derived normal and cancer cells instead of conventional cell lines for 3D engineering in vitro enables the development of more accurate 3D models, retaining the patient and tumor characteristics, and reflecting more appropriately tumor heterogeneity in the population. We highlighted, in this review, the paramount contributions of cell-secreted endogenous ECM components by focusing on the engineering of 3D cell models produced by the self-assembly approach of tissue engineering.
The 3D cell culture systems are increasingly gaining popularity in tumor and stem cell biology research. To date, numerous 3D cancer models have been specifically developed to take into account the in vivo architecture, tumor microenvironments,cell-cell and cell-surrounding matrix interactions, as well as signal transduction in cancer research. These models, ranging from simple 2D monoculture to complex bioengineered tissues, have been developed to better mimic the intrinsic discrepancies in the complexity and functionality of various tissues and tumors (Figure 6). These 3D systems also respectively harbor different intrinsic advantages and limitations and vary significantly in their biological relevance to the in vivo situation. More sophisticated 3D systems combining cancer and cells in self-secreted stroma could emphasize the importance of the tumor microenvironment and cell-cell crosstalk in cancer progression and invasiveness. Although each 3D culture technique/model is different in principle, each model shows their advantages and limitations. The selection of one model over another is therefore highly contextual and depends on the studied biological questions.
Among the numerous 3D models, we focused particularly on cancer models developed using the self-assembly method of tissue engineering. This method has been shown to overcome key limitations of currently using 2D flat monolayer cell culture and other simple 3D culture models, including uni-/multicellular spheroids, cancer-on-a-chip, organotypic slices of cancer tissues, and hydrogel- and scaffold-based systems. Indeed, the self-assembly method of tissue engineering allows the production of human/patient-derived 3D organ-specific tumors. As opposed to other 3D models, made with either natural (such as collagen, elastin, and glycosaminoglycans) or synthetic polymers to form various 3D structures, no exogenous scaffolding is needed. The self-assembly method of tissue engineering takes advantage of cell-specific self-synthesized ECM and assembly induced by ascorbate present in the culture media. This scaffold-free method is therefore not liable and not directly interfering with cell behavior, such as migration, proliferation, and aggregation influenced by the various mechanical properties and chemical composition of different bioscaffolding materials [241].
Other advantages of the scaffold-free self-assembly method of tissue engineering include cell-specific secretion of the tumor normoxic or hypoxic microenvironment favorable to cancer stemness through the generation of 3D tissues/tumors with diffusion-limited distribution of oxygen, nutrients, metabolites, and signaling molecules [242]. Enhanced cell survival through strong cell-cell contacts has also been observed using this approach [243]. Quite interestingly, gene-editing techniques such as CRISPR/Cas9 could also potentially be applied to this tissue-engineered approach to generate isogenic tissues to better control eventual experimental procedures.
Some important hurdles are however associated with this technique such as tedious isolation and purification procedures for specific cell types, and long production process and cell culture time requiring frequent changes of culture media by experienced laboratory staff to avoid contamination. Thereby, increased cost over the traditional culture model and large-scale production of the 3D tumor model generated by the self-assembly method could also become a disadvantage. Nevertheless, recent scientific breakthroughs have addressed these limitations using new cell culture protocols aimed at increasing the rate of ECM formation [244, 245]. Furthermore, although many questions and hurdles remain, biobanks of patient-derived 3D cancer models should further refine our understanding of interpatient as well as intrapatient heterogeneity, and hopefully, lead to personalized therapies for cancers.
Increasing the complexity of the microenvironment by creating multicellular niches is bound to further cancer research soon. With our expanding knowledge and control of the parameters required for optimal culture conditions for different cell types, models created in vitro will even be more realistic. Engineered 3D models will be useful tools to evaluate the impact of secreted molecules from specific cellular types on the normal and pathological functions. For most types of tumors, the incorporation of immune cells in tissue-engineered models represents one of the next important challenges. Another one consists of the incorporation of the vascular/lymphatic network simulating dynamic blood flow and providing the mechanical signals regulating tumor development and function. An increasing number of studies report the development of such perfusion systems applied to models of various primary and metastatic tumor types [20, 246–249].
Engineered cancer 3D models comprising patient-derived cells have also the potential of becoming valuable drug screening tools and are expected to significantly contribute to the progress of precision and personalized medicine. Indeed, the evidence is pointing out significant differences between treatment responses, partly due to intertumor variation and clonal variation within tumors [250]. New therapeutic approaches are therefore needed to improve current medical practices. Precision and personalized medicines are new concepts, often misused to describe what medicine should resemble in the future [251]. While precision medicine intends to target specific disease variants, personalized medicine is aimed at providing patient-tailored therapies. Examples of precision and personalized medicine include blood transfusion according to blood typing and autologous grafting, respectively. The 3D self-assembled tissues we discussed, which are entirely composed of human cells and matrix, display unique properties as a result of cell donor genetic and epigenetic backgrounds, lifestyle, and medical history. For instance, self-assembled tissues produced from cells of patients with pathological features can be used to evaluate drug efficacy/response in the context of precision medicine.
The 3D cell culture approaches hold a great promise and offer suitable systems for various purposes, ranging from disease modeling to drug target identification drug discovery, disease modeling, and cancer-targeted therapy. Continuous progress in tissue engineering, including the development of various bioreactor systems and 3D bioprinting, will improve the diversity, fidelity, and capacity of 3D culture models in cancer research. Besides the ability to generate geometric constructs containing viable cells, the 3D bioprinting technique may also facilitate high-throughput applications with precise reproducibility [88].
Acknowledgments
This work was supported by the Quebec Cell, Tissue, and Gene Therapy Network—ThéCell and the Vision Health Research Network (both thematic networks supported by the Fonds de recherche du Québec-Santé), the Association de la Neurofibromatose du Québec (ANFQ), Ferring Innovation, the Canadian Urological Oncology Group (CUOG), and the Fondation CHU de Québec. IL holds a Clinical Research Scholars-Junior 1 award from the Fonds de recherche du Québec-Santé (FRQ-S). FGL is the recipient of a tier-2 Canada Research Chair. LG is the recipient of a tier-1 Canada Research Chair. SL is a research scholar of the FRQ-S. LP is the recipient of a FRQ-S postdoctoral training award. We also thank Cassandra R. Goulet, Jennifer Bourland, Laure Gibot, Rémy Lamontagne, Aïcha Dede Djigo, Ioana Fugaru, and Julie Bérubé as well as the other students, postdoctoral fellows and research assistants for their helpful contributions to the development of the models. We gratefully acknowledge Dr. Nicolas Dupré for NF1 patient's recruitment and follow-up, Dr. Hélène T. Khuong for providing NF1-associated skin biopsies, and Dr. Carlos Lopez Valle for BCC biopsies.
Conflicts of Interest
The authors declare that they have no conflicts of interest.
Authors' Contributions
Vincent Roy, Brice Magne, François Gros-Louis, and Stéphane Bolduc contributed equally to this work.
References
- 1.Mak R. H., Hunt D., Shipley W. U., et al. Long-term outcomes in patients with muscle-invasive bladder cancer after selective bladder-preserving combined-modality therapy: a pooled analysis of Radiation Therapy Oncology Group protocols 8802, 8903, 9506, 9706, 9906, and 0233. Journal of Clinical Oncology. 2014;32(34):3801–3809. doi: 10.1200/JCO.2014.57.5548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Yuan Y., Wu L., Shen S., Wu S., Burdick M. M. Effect of alpha 2,6 sialylation on integrin-mediated adhesion of breast cancer cells to fibronectin and collagen IV. Life Sciences. 2016;149:138–145. doi: 10.1016/j.lfs.2016.02.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Michel M., L’Heureux N., Pouliot R., Xu W., Auger F. A., Germain L. Characterization of a new tissue-engineered human skin equivalent with hair. In Vitro Cellular & Developmental Biology - Animal. 1999;35(6):318–326. doi: 10.1007/s11626-999-0081-x. [DOI] [PubMed] [Google Scholar]
- 4.Huet E., Jaroz C., Nguyen H. Q., et al. Stroma in normal and cancer wound healing. The FEBS Journal. 2019;286(15):2909–2920. doi: 10.1111/febs.14842. [DOI] [PubMed] [Google Scholar]
- 5.Discher D. E., Janmey P., Wang Y.-l. Tissue cells feel and respond to the stiffness of their substrate. Science. 2005;310(5751):1139–1143. doi: 10.1126/science.1116995. [DOI] [PubMed] [Google Scholar]
- 6.Engler A. J., Sen S., Sweeney H. L., Discher D. E. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(4):677–689. doi: 10.1016/j.cell.2006.06.044. [DOI] [PubMed] [Google Scholar]
- 7.McKee T. J., Perlman G., Morris M., Komarova S. V. Extracellular matrix composition of connective tissues: a systematic review and meta-analysis. Scientific Reports. 2019;9(1, article 10542) doi: 10.1038/s41598-019-46896-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nystrom A., Bruckner-Tuderman L. Matrix molecules and skin biology. Seminars in Cell & Developmental Biology. 2019;89:136–146. doi: 10.1016/j.semcdb.2018.07.025. [DOI] [PubMed] [Google Scholar]
- 9.Lynch M. D., Watt F. M. Fibroblast heterogeneity: implications for human disease. Journal of Clinical Investigation. 2018;128(1):26–35. doi: 10.1172/JCI93555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen W., Frangogiannis N. G. Fibroblasts in post-infarction inflammation and cardiac repair. Biochim Biophys Acta. 2013;1833(4):945–953. doi: 10.1016/j.bbamcr.2012.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fukuda K., Ishida W., Fukushima A., Nishida T. Corneal fibroblasts as sentinel cells and local immune modulators in infectious keratitis. International Journal of Molecular Sciences. 2017;18(9, article 1831) doi: 10.3390/ijms18091831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ghetti M., Topouzi H., Theocharidis G., et al. Subpopulations of dermal skin fibroblasts secrete distinct extracellular matrix: implications for using skin substitutes in the clinic. British Journal of Dermatology. 2018;179:381–393. doi: 10.1111/bjd.16255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Klingberg F., Hinz B., White E. S. The myofibroblast matrix: implications for tissue repair and fibrosis. The Journal of Pathology. 2013;229(2):298–309. doi: 10.1002/path.4104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Blake K. J., Jiang X. R., Chiu I. M. Neuronal regulation of immunity in the skin and lungs. Trends in Neurosciences. 2019;42(8):537–551. doi: 10.1016/j.tins.2019.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Blais M., Mottier L., Germain M. A., Bellenfant S., Cadau S., Berthod F. Sensory neurons accelerate skin reepithelialization via substance P in an innervated tissue-engineered wound healing model. Tissue Engineering Part A. 2014;20(15-16):2180–2188. doi: 10.1089/ten.tea.2013.0535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dziki J. L., Wang D. S., Pineda C., Sicari B. M., Rausch T., Badylak S. F. Solubilized extracellular matrix bioscaffolds derived from diverse source tissues differentially influence macrophage phenotype. Journal of Biomedical Materials Research Part A. 2017;105(1):138–147. doi: 10.1002/jbm.a.35894. [DOI] [PubMed] [Google Scholar]
- 17.Richards C. D. Innate immune cytokines, fibroblast phenotypes, and regulation of extracellular matrix in lung. Journal of Interferon & Cytokine Research. 2017;37(2):52–61. doi: 10.1089/jir.2016.0112. [DOI] [PubMed] [Google Scholar]
- 18.Crowley T., Buckley C. D., Clark A. R. Stroma: the forgotten cells of innate immune memory. Clinical & Experimental Immunology. 2018;193(1):24–36. doi: 10.1111/cei.13149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bussard K. M., Mutkus L., Stumpf K., Gomez-Manzano C., Marini F. C. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Research. 2016;18(1):p. 84. doi: 10.1186/s13058-016-0740-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Yang Y., Yang X., Zou J., et al. Evaluation of photodynamic therapy efficiency using an in vitro three-dimensional microfluidic breast cancer tissue model. Lab on a Chip. 2015;15(3):735–744. doi: 10.1039/c4lc01065e. [DOI] [PubMed] [Google Scholar]
- 21.Bu L., Baba H., Yoshida N., et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38(25):4887–4901. doi: 10.1038/s41388-019-0765-y. [DOI] [PubMed] [Google Scholar]
- 22.Cirri P., Chiarugi P. Cancer associated fibroblasts: the dark side of the coin. American Journal of Cancer Research. 2011;1:482–497. [PMC free article] [PubMed] [Google Scholar]
- 23.Gieniec K. A., Butler L. M., Worthley D. L., Woods S. L. Cancer-associated fibroblasts--heroes or villains? British Journal of Cancer. 2019;121(4):293–302. doi: 10.1038/s41416-019-0509-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Orimo A., Gupta P. B., Sgroi D. C., et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–348. doi: 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
- 25.Chen X., Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nature Reviews Drug Discovery. 2019;18(2):99–115. doi: 10.1038/s41573-018-0004-1. [DOI] [PubMed] [Google Scholar]
- 26.Lambert A. W., Pattabiraman D. R., Weinberg R. A. Emerging biological principles of metastasis. Cell. 2017;168(4):670–691. doi: 10.1016/j.cell.2016.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hohenester E., Yurchenco P. D. Laminins in basement membrane assembly. Cell Adhesion & Migration. 2013;7(1):56–63. doi: 10.4161/cam.21831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pozzi A., Yurchenco P. D., Iozzo R. V. The nature and biology of basement membranes. Matrix Biology. 2017;57-58:1–11. doi: 10.1016/j.matbio.2016.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rousseau A., Fradette J., Bernard G., Gauvin R., Laterreur V., Bolduc S. Adipose-derived stromal cells for the reconstruction of a human vesical equivalent. Journal of Tissue Engineering and Regenerative Medicine. 2015;9(11):E135–E143. doi: 10.1002/term.1717. [DOI] [PubMed] [Google Scholar]
- 30.Theocharis A. D., Manou D., Karamanos N. K. The extracellular matrix as a multitasking player in disease. The FEBS Journal. 2019;286(15):2830–2869. doi: 10.1111/febs.14818. [DOI] [PubMed] [Google Scholar]
- 31.Adams J. C., Chiquet-Ehrismann R., Tucker R. P. The evolution of tenascins and fibronectin. Cell Adhesion & Migration. 2015;9(1-2):22–33. doi: 10.4161/19336918.2014.970030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Caon I., Bartolini B., Parnigoni A., et al. Revisiting the hallmarks of cancer: the role of hyaluronan. Seminars in Cancer Biology. 2019;62 doi: 10.1016/j.semcancer.2019.07.007. [DOI] [PubMed] [Google Scholar]
- 33.Iozzo R. V., Schaefer L. Proteoglycan form and function: a comprehensive nomenclature of proteoglycans. Matrix Biology. 2015;42:11–55. doi: 10.1016/j.matbio.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yuzhalin A. E., Lim S. Y., Kutikhin A. G., Gordon-Weeks A. N. Dynamic matrisome: ECM remodeling factors licensing cancer progression and metastasis. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 2018;1870(2):207–228. doi: 10.1016/j.bbcan.2018.09.002. [DOI] [PubMed] [Google Scholar]
- 35.Montagner M., Dupont S. Mechanical forces as determinants of disseminated metastatic cell fate. Cells. 2020;9(1):p. 250. doi: 10.3390/cells9010250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Quintero-Fabián S., Arreola R., Becerril-Villanueva E., et al. Role of matrix metalloproteinases in angiogenesis and cancer. Frontiers in oncology. 2019;9:p. 1370. doi: 10.3389/fonc.2019.01370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Simi A. K., Pang M. F., Nelson C. M. Extracellular matrix stiffness exists in a feedback loop that drives tumor progression. Advances in Experimental Medicine and Biology. 2018;1092:57–67. doi: 10.1007/978-3-319-95294-9_4. [DOI] [PubMed] [Google Scholar]
- 38.Eble J. A., Niland S. The extracellular matrix in tumor progression and metastasis. Clinical & Experimental Metastasis. 2019;36(3):171–198. doi: 10.1007/s10585-019-09966-1. [DOI] [PubMed] [Google Scholar]
- 39.Plotnikov S. V., Waterman C. M. Guiding cell migration by tugging. Current Opinion in Cell Biology. 2013;25(5):619–626. doi: 10.1016/j.ceb.2013.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Karamanou K., Franchi M., Vynios D., Brezillon S. Epithelial-to-mesenchymal transition and invadopodia markers in breast cancer: lumican a key regulator. Seminars in Cancer Biology. 2019;62 doi: 10.1016/j.semcancer.2019.08.003. [DOI] [PubMed] [Google Scholar]
- 41.Bland E., Dréau D., Burg K. J. L. Overcoming hypoxia to improve tissue-engineering approaches to regenerative medicine. Journal of Tissue Engineering and Regenerative Medicine. 2013;7(7):505–514. doi: 10.1002/term.540. [DOI] [PubMed] [Google Scholar]
- 42.Brown J. M., Wilson W. R. Exploiting tumour hypoxia in cancer treatment. Nature Reviews Cancer. 2004;4(6):437–447. doi: 10.1038/nrc1367. [DOI] [PubMed] [Google Scholar]
- 43.Hockel M., Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. JNCI Journal of the National Cancer Institute. 2001;93(4):266–276. doi: 10.1093/jnci/93.4.266. [DOI] [PubMed] [Google Scholar]
- 44.Harris A. L. Hypoxia -- a key regulatory factor in tumour growth. Nature Reviews Cancer. 2002;2(1):38–47. doi: 10.1038/nrc704. [DOI] [PubMed] [Google Scholar]
- 45.Wang G. L., Jiang B. H., Rue E. A., Semenza G. L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proceedings of the National Academy of Sciences. 1995;92(12):5510–5514. doi: 10.1073/pnas.92.12.5510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Carreau A., Hafny-Rahbi B. E., Matejuk A., Grillon C., Kieda C. Why is the partial oxygen pressure of human tissues a crucial parameter?Small molecules and hypoxia. Journal of Cellular and Molecular Medicine. 2011;15(6):1239–1253. doi: 10.1111/j.1582-4934.2011.01258.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Simon M. C., Keith B. The role of oxygen availability in embryonic development and stem cell function. Nature Reviews Molecular Cell Biology. 2008;9(4):285–296. doi: 10.1038/nrm2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Satyam A., Kumar P., Cigognini D., Pandit A., Zeugolis D. I. Low, but not too low, oxygen tension and macromolecular crowding accelerate extracellular matrix deposition in human dermal fibroblast culture. Acta Biomaterialia. 2016;44:221–231. doi: 10.1016/j.actbio.2016.08.008. [DOI] [PubMed] [Google Scholar]
- 49.Costa M., Pirraco R. P., Cerqueira M. T., Reis R. L., Marques A. P. Growth factor-free pre-vascularization of cell sheets for tissue engineering. Methods in Molecular Biology. 2016;1516:219–226. doi: 10.1007/7651_2016_362. [DOI] [PubMed] [Google Scholar]
- 50.Zhang L., Xing Q., Qian Z., et al. Hypoxia created human mesenchymal stem cell sheet for prevascularized 3D tissue construction. Advanced Healthcare Materials. 2016;5(3):342–352. doi: 10.1002/adhm.201500744. [DOI] [PubMed] [Google Scholar]
- 51.Yasui Y., Chijimatsu R., Hart D. A., et al. Preparation of scaffold-free tissue-engineered constructs derived from human synovial mesenchymal stem cells under low oxygen tension enhances their chondrogenic differentiation capacity. Tissue Engineering Part A. 2016;22(5-6):490–500. doi: 10.1089/ten.tea.2015.0458. [DOI] [PubMed] [Google Scholar]
- 52.Hudson K. D., Bonassar L. J. Hypoxic expansion of human mesenchymal stem cells enhances three-dimensional maturation of tissue-engineered intervertebral discs. Tissue Engineering Part A. 2017;23(7-8):293–300. doi: 10.1089/ten.TEA.2016.0270. [DOI] [PubMed] [Google Scholar]
- 53.Semenza G. L. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends in Pharmacological Sciences. 2012;33(4):207–214. doi: 10.1016/j.tips.2012.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wenger R. H., Stiehl D. P., Camenisch G. Integration of oxygen signaling at the consensus HRE. Science Signaling. 2005;2005(306, article re12) doi: 10.1126/stke.3062005re12. [DOI] [PubMed] [Google Scholar]
- 55.De Wever O., Mareel M. Role of tissue stroma in cancer cell invasion. The Journal of Pathology. 2003;200(4):429–447. doi: 10.1002/path.1398. [DOI] [PubMed] [Google Scholar]
- 56.Kapałczyńska M., Kolenda T., Przybyła W., et al. 2D and 3D cell cultures – a comparison of different types of cancer cell cultures. Archives of Medical Science. 2016;14:910–919. doi: 10.5114/aoms.2016.63743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Baker B. M., Chen C. S. Deconstructing the third dimension – how 3D culture microenvironments alter cellular cues. Journal of Cell Science. 2012;125(13):3015–3024. doi: 10.1242/jcs.079509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cukierman E., Pankov R., Stevens D. R., Yamada K. M. Taking cell-matrix adhesions to the third dimension. Science. 2001;294(5547):1708–1712. doi: 10.1126/science.1064829. [DOI] [PubMed] [Google Scholar]
- 59.Daley W. P., Peters S. B., Larsen M. Extracellular matrix dynamics in development and regenerative medicine. Journal of Cell Science. 2008;121(3):255–264. doi: 10.1242/jcs.006064. [DOI] [PubMed] [Google Scholar]
- 60.Petersen O. W., Ronnov-Jessen L., Howlett A. R., Bissell M. J. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proceedings of the National Academy of Sciences. 1992;89(19):9064–9068. doi: 10.1073/pnas.89.19.9064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.von der Mark K., Gauss V., von der Mark H., Muller P. Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature. 1977;267(5611):531–532. doi: 10.1038/267531a0. [DOI] [PubMed] [Google Scholar]
- 62.Gerlach J. C., Hout M., Edsbagge J., et al. Dynamic 3D culture promotes spontaneous embryonic stem cell differentiation in vitro. Tissue Engineering Part C: Methods. 2010;16(1):115–121. doi: 10.1089/ten.TEC.2008.0654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lei Y., Schaffer D. V. A fully defined and scalable 3D culture system for human pluripotent stem cell expansion and differentiation. Proceedings of the National Academy of Sciences. 2013;110(52):E5039–E5048. doi: 10.1073/pnas.1309408110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Birgersdotter A., Sandberg R., Ernberg I. Gene expression perturbation in vitro--A growing case for three-dimensional (3D) culture systems. Seminars in Cancer Biology. 2005;15(5):405–412. doi: 10.1016/j.semcancer.2005.06.009. [DOI] [PubMed] [Google Scholar]
- 65.Friedrich J., Seidel C., Ebner R., Kunz-Schughart L. A. Spheroid-based drug screen: considerations and practical approach. Nature Protocols. 2009;4(3):309–324. doi: 10.1038/nprot.2008.226. [DOI] [PubMed] [Google Scholar]
- 66.Eder T., Weber A., Neuwirt H., et al. Cancer-associated fibroblasts modify the response of prostate cancer cells to androgen and anti-androgens in three-dimensional spheroid culture. International Journal of Molecular Sciences. 2016;17(9):p. 1458. doi: 10.3390/ijms17091458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ghiabi P., Jiang J., Pasquier J., et al. Endothelial cells provide a notch-dependent pro-tumoral niche for enhancing breast cancer survival, stemness and pro-metastatic properties. PLoS One. 2014;9(11, article e112424) doi: 10.1371/journal.pone.0112424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Majety M., Pradel L. P., Gies M., Ries C. H. Fibroblasts influence survival and therapeutic response in a 3D co-culture model. PLoS One. 2015;10(6, article e0127948) doi: 10.1371/journal.pone.0127948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kenny H. A., Krausz T., Yamada S. D., Lengyel E. Use of a novel 3D culture model to elucidate the role of mesothelial cells, fibroblasts and extra-cellular matrices on adhesion and invasion of ovarian cancer cells to the omentum. International Journal of Cancer. 2007;121(7):1463–1472. doi: 10.1002/ijc.22874. [DOI] [PubMed] [Google Scholar]
- 70.Puls T. J., Tan X., Husain M., Whittington C. F., Fishel M. L., Voytik-Harbin S. L. Development of a novel 3D tumor-tissue invasion model for high-throughput, high-content phenotypic drug screening. Scientific Reports. 2018;8(1):p. 13039. doi: 10.1038/s41598-018-31138-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Loessner D., Stok K. S., Lutolf M. P., Hutmacher D. W., Clements J. A., Rizzi S. C. Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials. 2010;31(32):8494–8506. doi: 10.1016/j.biomaterials.2010.07.064. [DOI] [PubMed] [Google Scholar]
- 72.Ghosh S., Spagnoli G. C., Martin I., et al. Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study. Journal of Cellular Physiology. 2005;204(2):522–531. doi: 10.1002/jcp.20320. [DOI] [PubMed] [Google Scholar]
- 73.Qutub A. A., Popel A. S. Elongation, proliferation & migration differentiate endothelial cell phenotypes and determine capillary sprouting. BMC Systems Biology. 2009;3(1) doi: 10.1186/1752-0509-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chang T. T., Hughes-Fulford M. Monolayer and spheroid culture of human liver hepatocellular carcinoma cell line cells demonstrate distinct global gene expression patterns and functional phenotypes. Tissue Engineering Part A. 2009;15(3):559–567. doi: 10.1089/ten.tea.2007.0434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Longati P., Jia X., Eimer J., et al. 3D pancreatic carcinoma spheroids induce a matrix-rich, chemoresistant phenotype offering a better model for drug testing. BMC Cancer. 2013;13(1) doi: 10.1186/1471-2407-13-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Luca A. C., Mersch S., Deenen R., et al. Impact of the 3D microenvironment on phenotype, gene expression, and EGFR inhibition of colorectal cancer cell lines. PLoS One. 2013;8(3, article e59689) doi: 10.1371/journal.pone.0059689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Elliott N. T., Yuan F. A Review of Three-Dimensional In Vitro Tissue Models for Drug Discovery and Transport Studies. Journal of Pharmaceutical Sciences. 2011;100(1):59–74. doi: 10.1002/jps.22257. [DOI] [PubMed] [Google Scholar]
- 78.Beckwitt C. H., Clark A. M., Wheeler S., et al. Liver 'organ on a chip'. Experimental Cell Research. 2018;363(1):15–25. doi: 10.1016/j.yexcr.2017.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Krishnan V., Shuman L. A., Sosnoski D. M., Dhurjati R., Vogler E. A., Mastro A. M. Dynamic interaction between breast cancer cells and osteoblastic tissue: comparison of two- and three-dimensional cultures. Journal of Cellular Physiology. 2011;226(8):2150–2158. doi: 10.1002/jcp.22550. [DOI] [PubMed] [Google Scholar]
- 80.Katt M. E., Placone A. L., Wong A. D., Xu Z. S., Searson P. C. In vitro tumor models: advantages, disadvantages, variables, and selecting the right platform. Frontiers in Bioengineering and Biotechnology. 2016;4:p. 12. doi: 10.3389/fbioe.2016.00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Rodrigues T., Kundu B., Silva-Correia J., et al. Emerging tumor spheroids technologies for 3D in vitro cancer modeling. Pharmacology & Therapeutics. 2018;184, article S0163725817302681:201–211. doi: 10.1016/j.pharmthera.2017.10.018. [DOI] [PubMed] [Google Scholar]
- 82.Nath S., Devi G. R. Three-dimensional culture systems in cancer research: focus on tumor spheroid model. Pharmacology & Therapeutics. 2016;163:94–108. doi: 10.1016/j.pharmthera.2016.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lazzari G., Nicolas V., Matsusaki M., Akashi M., Couvreur P., Mura S. Multicellular spheroid based on a triple co-culture: a novel 3D model to mimic pancreatic tumor complexity. Acta Biomaterialia. 2018;78:296–307. doi: 10.1016/j.actbio.2018.08.008. [DOI] [PubMed] [Google Scholar]
- 84.Fang Y., Eglen R. M. Three-dimensional cell cultures in drug discovery and development. SLAS Discovery: Advancing the Science of Drug Discovery. 2017;22(5):456–472. doi: 10.1177/1087057117696795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Sant S., Johnston P. A. The production of 3D tumor spheroids for cancer drug discovery. Drug Discovery Today: Technologies. 2017;23:27–36. doi: 10.1016/j.ddtec.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kuo C. T., Wang J. Y., Lin Y. F., Wo A. M., Chen B. P. C., Lee H. Three-dimensional spheroid culture targeting versatile tissue bioassays using a PDMS-based hanging drop array. Scientific Reports. 2017;7(1, article 4363) doi: 10.1038/s41598-017-04718-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Blaeser A., Duarte Campos D. F., Puster U., Richtering W., Stevens M. M., Fischer H. Controlling shear stress in 3D bioprinting is a key factor to balance printing resolution and stem cell integrity. Advanced Healthcare Materials. 2016;5(3):326–333. doi: 10.1002/adhm.201500677. [DOI] [PubMed] [Google Scholar]
- 88.Knowlton S., Onal S., Yu C. H., Zhao J. J., Tasoglu S. Bioprinting for cancer research. Trends in Biotechnology. 2015;33(9):504–513. doi: 10.1016/j.tibtech.2015.06.007. [DOI] [PubMed] [Google Scholar]
- 89.Frantz C., Stewart K. M., Weaver V. M. The extracellular matrix at a glance. Journal of Cell Science. 2010;123(24):4195–4200. doi: 10.1242/jcs.023820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Auger F. A., Remy-Zolghadri M., Grenier G., Germain L. A truly new approach for tissue engineering: the LOEX self-assembly technique. Ernst Schering Res Found Workshop; 2002. [DOI] [PubMed] [Google Scholar]
- 91.Galbraith T., Roy V., Bourget J. M., et al. Cell seeding on UV-C-treated 3D polymeric templates allows for cost-effective production of small-caliber tissue-engineered blood vessels. Biotechnology Journal. 2019;14(1, article e1800306) doi: 10.1002/biot.201800306. [DOI] [PubMed] [Google Scholar]
- 92.L'heureux N., Pâquet S., Labbé R., Germain L., Auger F. A. A completely biological tissue-engineered human blood vessel. The FASEB Journal. 1998;12(1):47–56. doi: 10.1096/fasebj.12.1.47. [DOI] [PubMed] [Google Scholar]
- 93.Bouhout S., Perron É., Gauvin R., et al. In vitro reconstruction of an autologous, watertight, and resistant vesical equivalent. Tissue Engineering Part A. 2010;16(5):1539–1548. doi: 10.1089/ten.TEA.2009.0473. [DOI] [PubMed] [Google Scholar]
- 94.Vermette M., Trottier V., Menard V., Saintpierre L., Roy A., Fradette J. Production of a new tissue-engineered adipose substitute from human adipose- derived stromal cells. Biomaterials. 2007;28(18):2850–2860. doi: 10.1016/j.biomaterials.2007.02.030. [DOI] [PubMed] [Google Scholar]
- 95.Picard-Deland M., Ruel J., Galbraith T., et al. Tissue-engineered tubular heart valves combining a novel precontraction phase with the self-assembly method. Annals of Biomedical Engineering. 2017;45(2):427–438. doi: 10.1007/s10439-016-1708-1. [DOI] [PubMed] [Google Scholar]
- 96.Imbeault A., Bernard G., Rousseau A., et al. An endothelialized urothelial cell-seeded tubular graft for urethral replacement. Canadian Urological Association Journal. 2013;7(1-2):E4–E9. doi: 10.5489/cuaj.12217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Germain L., Auger F. A., Grandbois E., et al. Reconstructed human cornea produced in vitro by tissue engineering. Pathobiology. 1999;67(3):140–147. doi: 10.1159/000028064. [DOI] [PubMed] [Google Scholar]
- 98.Jean J., Lapointe M., Soucy J., Pouliot R. Development of an in vitro psoriatic skin model by tissue engineering. Journal of Dermatological Science. 2009;53(1):19–25. doi: 10.1016/j.jdermsci.2008.07.009. [DOI] [PubMed] [Google Scholar]
- 99.Bellemare J., Roberge C. J., Bergeron D., Lopez-Vallé C. A., Roy M., Moulin V. J. Epidermis promotes dermal fibrosis: role in the pathogenesis of hypertrophic scars. The Journal of Pathology. 2005;206(1):1–8. doi: 10.1002/path.1737. [DOI] [PubMed] [Google Scholar]
- 100.Paré B., Touzel-Deschênes L., Lamontagne R., et al. Early detection of structural abnormalities and cytoplasmic accumulation of TDP-43 in tissue-engineered skins derived from ALS patients. Acta Neuropathologica Communications. 2015;3(1):p. 5. doi: 10.1186/s40478-014-0181-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Proulx M., Mayrand D., Vincent C., et al. Short-term post-implantation dynamics of in vitro engineered human microvascularized adipose tissues. Biomedical Materials. 2018;13(6, article 065013) doi: 10.1088/1748-605X/aadff7. [DOI] [PubMed] [Google Scholar]
- 102.Bourland J., Fradette J., Auger F. A. Tissue-engineered 3D melanoma model with blood and lymphatic capillaries for drug development. Scientific Reports. 2018;8(1):p. 13191. doi: 10.1038/s41598-018-31502-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ringuette Goulet C., Bernard G., Chabaud S., et al. Tissue-engineered human 3D model of bladder cancer for invasion study and drug discovery. Biomaterials. 2017;145:233–241. doi: 10.1016/j.biomaterials.2017.08.041. [DOI] [PubMed] [Google Scholar]
- 104.Cortez Ghio S., Le-Bel G., Lavoie A., Larouche D., Germain L. Isolation and culture of human keratinocytes. Methods in Molecular Biology. 2019;1993:3–13. doi: 10.1007/978-1-4939-9473-1_1. [DOI] [PubMed] [Google Scholar]
- 105.Orabi H., Rousseau A., Laterreur V., Bolduc S. Optimization of the current self-assembled urinary bladder model: organ-specific stroma and smooth muscle inclusion. Canadian Urological Association Journal. 2015;9(9-10):599–607. doi: 10.5489/cuaj.2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Larouche D., Cantin-Warren L., Desgagné M., et al. Improved methods to produce tissue-engineered skin substitutes suitable for the permanent closure of full-thickness skin injuries. BioResearch Open Access. 2016;5(1):320–329. doi: 10.1089/biores.2016.0036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Saba I., Jakubowska W., Bolduc S., Chabaud S. Engineering tissues without the use of a synthetic scaffold: a twenty-year history of the self-assembly method. BioMed Research International. 2018;2018:13. doi: 10.1155/2018/5684679.5684679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bourland J., et al. Skin Tissue Engineering. New York: Humana; 2019. Isolation and culture of human dermal microvascular endothelial cells; pp. 79–90. [DOI] [PubMed] [Google Scholar]
- 109.Berthod F., Symes J., Tremblay N., Medin J. A., Auger F. A. Spontaneous fibroblast-derived pericyte recruitment in a human tissue-engineered angiogenesis model in vitro. Journal of Cellular Physiology. 2012;227(5):2130–2137. doi: 10.1002/jcp.22943. [DOI] [PubMed] [Google Scholar]
- 110.Rochon M. H., Fradette J., Fortin V., et al. Normal human epithelial cells regulate the size and morphology of tissue-engineered capillaries. Tissue Engineering Part A. 2010;16(5):1457–1468. doi: 10.1089/ten.TEA.2009.0090. [DOI] [PubMed] [Google Scholar]
- 111.Gibot L., Galbraith T., Huot J., Auger F. A. A preexisting microvascular network benefits in vivo revascularization of a microvascularized tissue-engineered skin substitute. Tissue Engineering Part A. 2010;16(10):3199–3206. doi: 10.1089/ten.TEA.2010.0189. [DOI] [PubMed] [Google Scholar]
- 112.Guillemette M. D., Gauvin R., Perron C., Labbé R., Germain L., Auger F. A. Tissue-engineered vascular adventitia with vasa vasorum improves graft integration and vascularization through inosculation. Tissue Engineering Part A. 2010;16(8):2617–2626. doi: 10.1089/ten.tea.2009.0612. [DOI] [PubMed] [Google Scholar]
- 113.Tremblay P. L., Huot J., Auger F. A. Mechanisms by which E-selectin regulates diapedesis of colon cancer cells under flow conditions. Cancer Research. 2008;68(13):5167–5176. doi: 10.1158/0008-5472.CAN-08-1229. [DOI] [PubMed] [Google Scholar]
- 114.Chabaud S., Rousseau A., Marcoux T. L., Bolduc S. Inexpensive production of near-native engineered stromas. Journal of Tissue Engineering and Regenerative Medicine. 2017;11(5):1377–1389. doi: 10.1002/term.2036. [DOI] [PubMed] [Google Scholar]
- 115.Gibot L., Galbraith T., Kloos B., et al. Cell-based approach for 3D reconstruction of lymphatic capillaries in vitro reveals distinct functions of HGF and VEGF-C in lymphangiogenesis. Biomaterials. 2016;78:129–139. doi: 10.1016/j.biomaterials.2015.11.027. [DOI] [PubMed] [Google Scholar]
- 116.Gibot L., Galbraith T., Bourland J., Rogic A., Skobe M., Auger F. A. Tissue-engineered 3D human lymphatic microvascular network for in vitro studies of lymphangiogenesis. Nature Protocols. 2017;12(5):1077–1088. doi: 10.1038/nprot.2017.025. [DOI] [PubMed] [Google Scholar]
- 117.Erhard H., Rietveld F. J. R., van Altena M. C., Bröcker E. B., Ruiter D. J., de Waal R. M. W. Transition of horizontal to vertical growth phase melanoma is accompanied by induction of vascular endothelial growth factor expression and angiogenesis. Melanoma Research. 1997;7(Supplement 2):p. S27. doi: 10.1097/00008390-199708001-00005. [DOI] [PubMed] [Google Scholar]
- 118.Lobb R. J., Lima L. G., Moller A. Exosomes: key mediators of metastasis and pre-metastatic niche formation. Seminars in Cell & Developmental Biology. 2017;67:3–10. doi: 10.1016/j.semcdb.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 119.Narkhede A. A., Shevde L. A., Rao S. S. Biomimetic strategies to recapitulate organ specific microenvironments for studying breast cancer metastasis. International Journal of Cancer. 2017;141(6):1091–1109. doi: 10.1002/ijc.30748. [DOI] [PubMed] [Google Scholar]
- 120.Cazzaniga W., Nebuloni M., Longhi E., et al. Human Prostate Tissue-derived Extracellular Matrix as a Model of Prostate Microenvironment. European Urology Focus. 2016;2(4):400–408. doi: 10.1016/j.euf.2016.02.016. [DOI] [PubMed] [Google Scholar]
- 121.Tian X., Werner M. E., Roche K. C., et al. Organ-specific metastases obtained by culturing colorectal cancer cells on tissue-specific decellularized scaffolds. Nature Biomedical Engineering. 2018;2(6):443–452. doi: 10.1038/s41551-018-0231-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Xiong G., Flynn T. J., Chen J., Trinkle C., Xu R. Development of anex vivobreast cancer lung colonization model utilizing a decellularized lung matrix. Integrative Biology: Quantitative Biosciences from Nano to Macro. 2015;7(12):1518–1525. doi: 10.1039/c5ib00157a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Contag C. H., Lie W. R., Bammer M. C., et al. Monitoring dynamic interactions between breast cancer cells and human bone tissue in a co-culture model. Molecular Imaging and Biology. 2014;16(2):158–166. doi: 10.1007/s11307-013-0685-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pathi S. P., Kowalczewski C., Tadipatri R., Fischbach C. A novel 3-D mineralized tumor model to study breast cancer bone metastasis. PLoS One. 2010;5(1, article e8849) doi: 10.1371/journal.pone.0008849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Huang R., Rofstad E. K. Integrins as therapeutic targets in the organ-specific metastasis of human malignant melanoma. Journal of Experimental & Clinical Cancer Research. 2018;37(1):p. 92. doi: 10.1186/s13046-018-0763-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Carrier P., Deschambeault A., Audet C., et al. Impact of cell source on human cornea reconstructed by tissue engineering. Investigative Ophthalmology & Visual Science. 2009;50(6):2645–2652. doi: 10.1167/iovs.08-2001. [DOI] [PubMed] [Google Scholar]
- 127.Bouhout S., Chabaud S., Bolduc S. Organ-specific matrix self-assembled by mesenchymal cells improves the normal urothelial differentiation in vitro. World Journal of Urology. 2016;34(1):121–130. doi: 10.1007/s00345-015-1596-2. [DOI] [PubMed] [Google Scholar]
- 128.Corriveau M. P., Boufaied I., Lessard J., et al. The fibrotic phenotype of systemic sclerosis fibroblasts varies with disease duration and severity of skin involvement: reconstitution of skin fibrosis development using a tissue engineering approach. The Journal of Pathology. 2009;217(4):534–542. doi: 10.1002/path.2482. [DOI] [PubMed] [Google Scholar]
- 129.Cameron M. C., Lee E., Hibler B. P., et al. Basal cell carcinoma: epidemiology; pathophysiology; clinical and histological subtypes; and disease associations. Journal of the American Academy of Dermatology. 2019;80(2):303–317. doi: 10.1016/j.jaad.2018.03.060. [DOI] [PubMed] [Google Scholar]
- 130.Goldberg L. H. Basal cell carcinoma. The Lancet. 1996;347(9002):663–667. doi: 10.1016/S0140-6736(96)91208-8. [DOI] [PubMed] [Google Scholar]
- 131.Rubin A. I., Chen E. H., Ratner D. Basal-cell carcinoma. New England Journal of Medicine. 2005;353(21):2262–2269. doi: 10.1056/NEJMra044151. [DOI] [PubMed] [Google Scholar]
- 132.Mackiewicz-Wysocka M., Bowszyc-Dmochowska M., Strzelecka-Weklar D., Danczak-Pazdrowska A., Adamski Z. Basal cell carcinoma—diagnosis. Contemporary Oncology. 2013;4(4):337–342. doi: 10.5114/wo.2013.35684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.de Vries E., Louwman M., Bastiaens M., de Gruijl F., Coebergh J. W. Rapid and continuous increases in incidence rates of basal cell carcinoma in the Southeast Netherlands since 1973. Journal of Investigative Dermatology. 2004;123(4):634–638. doi: 10.1111/j.0022-202X.2004.23306.x. [DOI] [PubMed] [Google Scholar]
- 134.Rudolph C., Schnoor M., Eisemann N., Katalinic A. Incidence trends of nonmelanoma skin cancer in Germany from 1998 to 2010. JDDG: Journal der Deutschen Dermatologischen Gesellschaft. 2015;13(8):788–797. doi: 10.1111/ddg.12690. [DOI] [PubMed] [Google Scholar]
- 135.Staples M. P., Elwood M., Burton R. C., Williams J. L., Marks R., Giles G. G. Non-melanoma skin cancer in Australia: the 2002 national survey and trends since 1985. Medical Journal of Australia. 2006;184(1):6–10. doi: 10.5694/j.1326-5377.2006.tb00086.x. [DOI] [PubMed] [Google Scholar]
- 136.Sng J., Koh D., Siong W. C., Choo T. B. Skin cancer trends among Asians living in Singapore from 1968 to 2006. Journal of the American Academy of Dermatology. 2009;61(3):426–432. doi: 10.1016/j.jaad.2009.03.031. [DOI] [PubMed] [Google Scholar]
- 137.Demers A. A., Nugent Z., Mihalcioiu C., Wiseman M. C., Kliewer E. V. Trends of nonmelanoma skin cancer from 1960 through 2000 in a Canadian population. Journal of the American Academy of Dermatology. 2005;53(2):320–328. doi: 10.1016/j.jaad.2005.03.043. [DOI] [PubMed] [Google Scholar]
- 138.Wu S., Han J., Li W.-Q., Li T., Qureshi A. A. Basal-cell carcinoma incidence and associated risk factors in U.S. women and men. American Journal of Epidemiology. 2013;178(6):890–897. doi: 10.1093/aje/kwt073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rogers H. W., Coldiron B. M. A relative value unit-based cost comparison of treatment modalities for nonmelanoma skin cancer: effect of the loss of the Mohs multiple surgery reduction exemption. Journal of the American Academy of Dermatology. 2009;61(1):96–103. doi: 10.1016/j.jaad.2008.07.047. [DOI] [PubMed] [Google Scholar]
- 140.Gorlin R. J., Goltz R. W. Multiple nevoid basal-cell Epithelioma, jaw cysts and bifid Rib. New England Journal of Medicine. 1960;262(18):908–912. doi: 10.1056/NEJM196005052621803. [DOI] [PubMed] [Google Scholar]
- 141.Hahn H., Wicking C., Zaphiropoulos P. G., et al. Mutations of the Human Homolog of Drosophila _patched_ in the Nevoid Basal Cell Carcinoma Syndrome. Cell. 1996;85(6):841–851. doi: 10.1016/S0092-8674(00)81268-4. [DOI] [PubMed] [Google Scholar]
- 142.Johnson R. L., Rothman A. L., Xie J., et al. Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science. 1996;272(5268):1668–1671. doi: 10.1126/science.272.5268.1668. [DOI] [PubMed] [Google Scholar]
- 143.Oro A. E., Higgins K. M., Hu Z., Bonifas J. M., Epstein EH Jr, Scott M. P. Basal cell carcinomas in mice overexpressing sonic hedgehog. Science. 1997;276(5313):817–821. doi: 10.1126/science.276.5313.817. [DOI] [PubMed] [Google Scholar]
- 144.Xie J., Murone M., Luoh S. M., et al. Activating Smoothened mutations in sporadic basal-cell carcinoma. Nature. 1998;391(6662):90–92. doi: 10.1038/34201. [DOI] [PubMed] [Google Scholar]
- 145.Nilsson M., Undèn A. B., Krause D., et al. Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proceedings of the National Academy of Sciences. 2000;97(7):3438–3443. doi: 10.1073/pnas.050467397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Grachtchouk M., Mo R., Yu S., et al. Basal cell carcinomas in mice overexpressing Gli2 in skin. Nature Genetics. 2000;24(3):216–217. doi: 10.1038/73417. [DOI] [PubMed] [Google Scholar]
- 147.Aszterbaum M., Epstein J., Oro A., et al. Ultraviolet and ionizing radiation enhance the growth of BCCs and trichoblastomas in patched heterozygous knockout mice. Nature Medicine. 1999;5(11):1285–1291. doi: 10.1038/15242. [DOI] [PubMed] [Google Scholar]
- 148.Tang J. Y., Mackay-Wiggan J. M., Aszterbaum M., et al. Inhibiting the hedgehog pathway in patients with the basal-cell nevus syndrome. New England Journal of Medicine. 2002;366:2180–2188. doi: 10.1056/NEJMoa1113538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Auepemkiate S., Boonyaphiphat P., Thongsuksai P. p53 expression related to the aggressive infiltrative histopathological feature of basal cell carcinoma. Histopathology. 2002;40(6):568–573. doi: 10.1046/j.1365-2559.2002.01393.x. [DOI] [PubMed] [Google Scholar]
- 150.Yang S. H., Andl T., Grachtchouk V., et al. Pathological responses to oncogenic Hedgehog signaling in skin are dependent on canonical Wnt/β-catenin signaling. Nature Genetics. 2008;40(9):1130–1135. doi: 10.1038/ng.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Eberl M., Klingler S., Mangelberger D., et al. Hedgehog-EGFR cooperation response genes determine the oncogenic phenotype of basal cell carcinoma and tumour-initiating pancreatic cancer cells. EMBO Molecular Medicine. 2012;4(3):218–233. doi: 10.1002/emmm.201100201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Sneddon J. B., Zhen H. H., Montgomery K., et al. Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proceedings of the National Academy of Sciences. 2006;103(40):14842–14847. doi: 10.1073/pnas.0606857103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Lacina L., Smetana K., Dvořánková B., et al. Stromal fibroblasts from basal cell carcinoma affect phenotype of normal keratinocytes. British Journal of Dermatology. 2007;156(5):819–829. doi: 10.1111/j.1365-2133.2006.07728.x. [DOI] [PubMed] [Google Scholar]
- 154.Williams J. A., Guicherit O. M., Zaharian B. I., et al. Identification of a small molecule inhibitor of the hedgehog signaling pathway: effects on basal cell carcinoma-like lesions. Proceedings of the National Academy of Sciences. 2003;100(8):4616–4621. doi: 10.1073/pnas.0732813100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Williams V. C., Lucas J., Babcock M. A., Gutmann D. H., Korf B., Maria B. L. Neurofibromatosis type 1 revisited. Pediatrics. 2009;123(1):124–133. doi: 10.1542/peds.2007-3204. [DOI] [PubMed] [Google Scholar]
- 156.Ballester R., Marchuk D., Boguski M., et al. The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell. 1990;63(4):851–859. doi: 10.1016/0092-8674(90)90151-4. [DOI] [PubMed] [Google Scholar]
- 157.Xu G., Lin B., Tanaka K., et al. The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell. 1990;63(4):835–841. doi: 10.1016/0092-8674(90)90149-9. [DOI] [PubMed] [Google Scholar]
- 158.Martin G. A., Viskoohil D., Bollag G., et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990;63(4):843–849. doi: 10.1016/0092-8674(90)90150-d. [DOI] [PubMed] [Google Scholar]
- 159.Ferner R. E. Neurofibromatosis 1 and neurofibromatosis 2: a twenty first century perspective. The Lancet Neurology. 2007;6(4):340–351. doi: 10.1016/S1474-4422(07)70075-3. [DOI] [PubMed] [Google Scholar]
- 160.Ferner R. E., Gutmann D. H. International consensus statement on malignant peripheral nerve sheath tumors in neurofibromatosis. Cancer Research. 2002;62(5):1573–1577. [PubMed] [Google Scholar]
- 161.Ortonne N., Wolkenstein P., Blakeley J. O., et al. Cutaneous neurofibromas. Neurology. 2018;91(2) Supplement 1:S5–S13. doi: 10.1212/WNL.0000000000005792. [DOI] [PubMed] [Google Scholar]
- 162.Jo V. Y., Fletcher C. D. M. WHO classification of soft tissue tumours: an update based on the 2013 (4th) edition. Pathology. 2014;46(2):95–104. doi: 10.1097/PAT.0000000000000050. [DOI] [PubMed] [Google Scholar]
- 163.Staser K., Yang F. C., Clapp D. W. Mast cells and the neurofibroma microenvironment. Blood. 2010;116(2):157–164. doi: 10.1182/blood-2009-09-242875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Tonsgard J. H. Clinical manifestations and management of neurofibromatosis type 1. Seminars in Pediatric Neurology. 2006;13(1):2–7. doi: 10.1016/j.spen.2006.01.005. [DOI] [PubMed] [Google Scholar]
- 165.Garcia-Linares C., Fernández-Rodríguez J., Terribas E., et al. Dissecting loss of heterozygosity (LOH) in neurofibromatosis type 1-associated neurofibromas: importance of copy neutral LOH. Human Mutation. 2011;32(1):78–90. doi: 10.1002/humu.21387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Serra E., Puig S., Otero D., et al. Confirmation of a Double-Hit Model for the _NF1_ Gene in Benign Neurofibromas. The American Journal of Human Genetics. 1997;61(3):512–519. doi: 10.1086/515504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Cannon A., Chen M. J., Li P., et al. Cutaneous neurofibromas in neurofibromatosis type I: a quantitative natural history study. Orphanet Journal of Rare Diseases. 2018;13(1):p. 31. doi: 10.1186/s13023-018-0772-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Pasmant E., Vidaud M., Vidaud D., Wolkenstein P. Neurofibromatosis type 1: from genotype to phenotype. Journal of Medical Genetics. 2012;49(8):483–489. doi: 10.1136/jmedgenet-2012-100978. [DOI] [PubMed] [Google Scholar]
- 169.Isakson S. H., Rizzardi A. E., Coutts A. W., et al. Genetically engineered minipigs model the major clinical features of human neurofibromatosis type 1. Communications Biology. 2018;1(1):p. 158. doi: 10.1038/s42003-018-0163-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Chen Z., Mo J., Brosseau J. P., et al. Spatiotemporal loss ofNF1in Schwann cell lineage leads to different types of cutaneous neurofibroma susceptible to modification by the Hippo pathway. Cancer Discovery. 2019;9(1):114–129. doi: 10.1158/2159-8290.CD-18-0151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Zhu Y., Ghosh P., Charnay P., Burns D. K., Parada L. F. Neurofibromas in NF1: Schwann cell origin and role of tumor environment. Science. 2002;296(5569):920–922. doi: 10.1126/science.1068452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Kraniak J. M., Chalasani A., Wallace M. R., Mattingly R. R. Development of 3D culture models of plexiform neurofibroma and initial application for phenotypic characterization and drug screening. Experimental Neurology. 2018;299(Part B):289–298. doi: 10.1016/j.expneurol.2017.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Carrió M., Mazuelas H., Richaud-Patin Y., et al. Reprogramming captures the genetic and tumorigenic properties of neurofibromatosis type 1 plexiform neurofibromas. Stem Cell Reports. 2019;12(2):411–426. doi: 10.1016/j.stemcr.2019.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Roy V., Touzel-Deschênes L., Marques E., et al. Tissue-engineered skin model derived from neurofibromatosis type 1 (NF1) patients to study tumor genesis and to predict response to therapy (P4.057) Neurology. 2017;88 [Google Scholar]
- 175.Karimkhani C., Green A. C., Nijsten T., et al. The global burden of melanoma: results from the Global Burden of Disease Study 2015. British Journal of Dermatology. 2017;177(1):134–140. doi: 10.1111/bjd.15510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Howlader N., Krapcho N. A., Miller D., et al. SEER cancer statistics review—cancer stat facts: melanoma of the skin. 2019, https://seer.cancer.gov/statfacts/html/melan.html.
- 177.Miller K. D., Nogueira L., Mariotto A. B., et al. Cancer treatment and survivorship statistics, 2019. CA: A Cancer Journal for Clinicians. 2019;69(5):363–385. doi: 10.3322/caac.21565. [DOI] [PubMed] [Google Scholar]
- 178.MCGOVERN V. J., MACKIE B. S. The relationship of solar radiation to melanoblastoma. ANZ Journal of Surgery. 1959;28(4):257–262. doi: 10.1111/j.1445-2197.1959.tb06967.x. [DOI] [PubMed] [Google Scholar]
- 179.Garibyan L., Fisher D. E. How sunlight causes melanoma. Current Oncology Reports. 2010;12(5):319–326. doi: 10.1007/s11912-010-0119-y. [DOI] [PubMed] [Google Scholar]
- 180.Trucco L. D., Mundra P. A., Hogan K., et al. Ultraviolet radiation-induced DNA damage is prognostic for outcome in melanoma. Nature Medicine. 2019;25(2):221–224. doi: 10.1038/s41591-018-0265-6. [DOI] [PubMed] [Google Scholar]
- 181.Li F. Z., Dhillon A. S., Anderson R. L., McArthur G., Ferrao P. T. Phenotype switching in melanoma: implications for progression and therapy. Frontiers in Oncology. 2015;5:p. 31. doi: 10.3389/fonc.2015.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Itakura E., Huang R.-R., Wen D.-R., Paul E., Wünsch P. H., Cochran A. J. IL-10 expression by primary tumor cells correlates with melanoma progression from radial to vertical growth phase and development of metastatic competence. Modern Pathology. 2011;24(6):801–809. doi: 10.1038/modpathol.2011.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Marconi A., Quadri M., Saltari A., Pincelli C. Progress in melanoma modelling in vitro. Experimental Dermatology. 2018;27(5):578–586. doi: 10.1111/exd.13670. [DOI] [PubMed] [Google Scholar]
- 184.Eves P., Layton C., Hedley S., et al. Characterization of an in vitro model of human melanoma invasion based on reconstructed human skin. British Journal of Dermatology. 2000;142(2):210–222. doi: 10.1046/j.1365-2133.2000.03287.x. [DOI] [PubMed] [Google Scholar]
- 185.Hill D. S., Robinson N. D. P., Caley M. P., et al. A novel fully humanized 3D skin equivalent to model early melanoma invasion. Molecular Cancer Therapeutics. 2015;14(11):2665–2673. doi: 10.1158/1535-7163.Mct-15-0394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Morales D., Lombart F., Truchot A., et al. 3D coculture models underline metastatic melanoma cell sensitivity to vemurafenib. Tissue Engineering Part A. 2019;25(15-16):1116–1126. doi: 10.1089/ten.TEA.2018.0210. [DOI] [PubMed] [Google Scholar]
- 187.Shah M. K., Leary E. A., Darling E. M. Integration of hyper-compliant microparticles into a 3D melanoma tumor model. Journal of Biomechanics. 2019;82:46–53. doi: 10.1016/j.jbiomech.2018.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Vörsmann H., Groeber F., Walles H., et al. Development of a human three-dimensional organotypic skin-melanoma spheroid model for in vitro drug testing. Cell Death & Disease. 2013;4(7, article e719) doi: 10.1038/cddis.2013.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Gibot L., Galbraith T., Huot J., Auger F. A. Development of a tridimensional microvascularized human skin substitute to study melanoma biology. Clinical & Experimental Metastasis. 2013;30(1):83–90. doi: 10.1007/s10585-012-9511-3. [DOI] [PubMed] [Google Scholar]
- 190.Bray F., Ferlay J., Soerjomataram I., Siegel R. L., Torre L. A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 191.Edgren G., Liang L., Adami H. O., Chang E. T. Enigmatic sex disparities in cancer incidence. European Journal of Epidemiology. 2012;27(3):187–196. doi: 10.1007/s10654-011-9647-5. [DOI] [PubMed] [Google Scholar]
- 192.Sanli O., Dobruch J., Knowles M. A., et al. Bladder cancer. Nature Reviews Disease Primers. 2017;3(1, article 17022) doi: 10.1038/nrdp.2017.22. [DOI] [PubMed] [Google Scholar]
- 193.Cao M., Tasian G., Wang M. H., Liu B., Cunha G., Baskin L. Urothelium-derived Sonic hedgehog promotes mesenchymal proliferation and induces bladder smooth muscle differentiation. Differentiation. 2010;79(4-5):244–250. doi: 10.1016/j.diff.2010.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Tasian G., Cunha G., Baskin L. Smooth muscle differentiation and patterning in the urinary bladder. Differentiation. 2010;80(2-3):106–117. doi: 10.1016/j.diff.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Mendelsohn C. Going in circles: conserved mechanisms control radial patterning in the urinary and digestive tracts. Journal of Clinical Investigation. 2006;116(3):635–637. doi: 10.1172/JCI27985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Baskin L. S., Hayward S. W., Young P., Cunha G. R. Role of mesenchymal-epithelial interactions in normal bladder development. Journal of Urology. 1996;156(5):1820–1827. doi: 10.1016/S0022-5347(01)65545-9. [DOI] [PubMed] [Google Scholar]
- 197.Vasyutin I., Zerihun L., Ivan C., Atala A. Bladder organoids and spheroids: potential tools for normal and diseased tissue modelling. Anticancer Research. 2019;39(3):1105–1118. doi: 10.21873/anticanres.13219. [DOI] [PubMed] [Google Scholar]
- 198.Mullenders J., de Jongh E., Brousali A., et al. Mouse and human urothelial cancer organoids: a tool for bladder cancer research. Proceedings of the National Academy of Sciences. 2019;116(10):4567–4574. doi: 10.1073/pnas.1803595116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Yoshida T., Singh A. K., Bishai W. R., McConkey D. J., Bivalacqua T. J. Organoid culture of bladder cancer cells. Investigative and Clinical Urology. 2018;59(3):149–151. doi: 10.4111/icu.2018.59.3.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Yoshida T., Sopko N., Kates M., et al. Impact of spheroid culture on molecular and functional characteristics of bladder cancer cell lines. Oncology Letters. 2019;18:4923–4929. doi: 10.3892/ol.2019.10786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Amaral R. L. F., Miranda M., Marcato P. D., Swiech K. Comparative analysis of 3D bladder tumor spheroids obtained by forced floating and hanging drop methods for drug screening. Frontiers in Physiology. 2017;8:p. 605. doi: 10.3389/fphys.2017.00605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Frandsen S. K., Gibot L., Madi M., Gehl J., Rols M. P. Calcium electroporation: evidence for differential effects in normal and malignant cell lines, evaluated in a 3D spheroid model. PLoS One. 2015;10(12, e0144028) doi: 10.1371/journal.pone.0144028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Yoshida T., Sopko N. A., Kates M., et al. Three-dimensional organoid culture reveals involvement of Wnt/β-catenin pathway in proliferation of bladder cancer cells. Oncotarget. 2018;9(13):11060–11070. doi: 10.18632/oncotarget.24308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Lee S. H., Hu W., Matulay J. T., et al. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 2018;173(2):515–528.e17. doi: 10.1016/j.cell.2018.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Yoshida T., Okuyama H., Endo H., Inoue M. Spheroid cultures of primary urothelial cancer cells: cancer tissue-originated spheroid (CTOS) method. Methods in Molecular Biology. 2018;1655:145–153. doi: 10.1007/978-1-4939-7234-0_12. [DOI] [PubMed] [Google Scholar]
- 206.Namekawa T., et al. ALDH1A1 in patient-derived bladder cancer spheroids activates retinoic acid signaling leading to TUBB3 overexpression and tumor progression. Int J Cancer. 2020;146:1099–1113. doi: 10.1002/ijc.32505. [DOI] [PubMed] [Google Scholar]
- 207.Landreville S., Agapova O. A., Harbour J. W. Emerging insights into the molecular pathogenesis of uveal melanoma. Future Oncology. 2008;4(5):629–636. doi: 10.2217/14796694.4.5.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Rodrigues M., de Koning L., Coupland S., et al. So close, yet so far: discrepancies between uveal and other melanomas. A position paper from UM Cure 2020. Cancers. 2019;11(7):p. 1032. doi: 10.3390/cancers11071032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Singh A. D., Turell M. E., Topham A. K. Uveal melanoma: trends in incidence, treatment, and survival. Ophthalmology. 2011;118(9):1881–1885. doi: 10.1016/j.ophtha.2011.01.040. [DOI] [PubMed] [Google Scholar]
- 210.Nayman T., Bostan C., Logan P., Burnier M. N., Jr. Uveal melanoma risk factors: a systematic review of meta-analyses. Current Eye Research. 2017;42(8):1085–1093. doi: 10.1080/02713683.2017.1297997. [DOI] [PubMed] [Google Scholar]
- 211.Singh A. D., Shields C. L., de Potter P., et al. Familial uveal melanoma. Clinical observations on 56 patients. Archives of Ophthalmology. 1996;114(4):392–399. doi: 10.1001/archopht.1996.01100130388005. [DOI] [PubMed] [Google Scholar]
- 212.Repo P., Järvinen R. S., Jäntti J. E., et al. Population-based analysis of BAP1 germline variations in patients with uveal melanoma. Human Molecular Genetics. 2019;28(14):2415–2426. doi: 10.1093/hmg/ddz076. [DOI] [PubMed] [Google Scholar]
- 213.Park J. J., Diefenbach R. J., Joshua A. M., Kefford R. F., Carlino M. S., Rizos H. Oncogenic signaling in uveal melanoma. Pigment Cell & Melanoma Research. 2018;31(6):661–672. doi: 10.1111/pcmr.12708. [DOI] [PubMed] [Google Scholar]
- 214.Kaliki S., Shields C. L. Uveal melanoma: relatively rare but deadly cancer. Eye. 2017;31(2):241–257. doi: 10.1038/eye.2016.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Wilson M. W., Hungerford J. L. Comparison of episcleral plaque and proton beam radiation therapy for the treatment of choroidal melanoma. Ophthalmology. 1999;106(8):1579–1587. doi: 10.1016/S0161-6420(99)90456-6. [DOI] [PubMed] [Google Scholar]
- 216.Rossi E., Schinzari G., Zizzari I. G., et al. Immunological backbone of uveal melanoma: is there a rationale for immunotherapy? Cancers. 2019;11(8):p. 1055. doi: 10.3390/cancers11081055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Schank T. E., Hassel J. C. Immunotherapies for the treatment of uveal melanoma-history and future. Cancers. 2019;11(8):p. 1048. doi: 10.3390/cancers11081048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Weidmann C., Pomerleau J., Trudel-Vandal L., Landreville S. Differential responses of choroidal melanocytes and uveal melanoma cells to low oxygen conditions. Molecular Vision. 2017;23:103–115. [PMC free article] [PubMed] [Google Scholar]
- 219.Djigo A. D., Bérubé J., Landreville S., Proulx S. Characterization of a tissue-engineered choroid. Acta Biomaterialia. 2019;84:305–316. doi: 10.1016/j.actbio.2018.11.033. [DOI] [PubMed] [Google Scholar]
- 220.Komez A., Baran E. T., Erdem U., Hasirci N., Hasirci V. Construction of a patterned hydrogel-fibrous mat bilayer structure to mimic choroid and Bruch’s membrane layers of retina. Journal of Biomedical Materials Research Part A. 2016;104(9):2166–2177. doi: 10.1002/jbm.a.35756. [DOI] [PubMed] [Google Scholar]
- 221.Shokoohmand A., Jeon J. E., Theodoropoulos C., Baldwin J. G., Hutmacher D. W., Feigl B. A novel 3D cultured model for studying early changes in age-related macular degeneration. Macromolecular Bioscience. 2017;17(12) doi: 10.1002/mabi.201700221. [DOI] [PubMed] [Google Scholar]
- 222.Weidmann C., Bérubé J., Piquet L., de la Fouchardière A., Landreville S. Expression of the serotonin receptor 2B in uveal melanoma and effects of an antagonist on cell lines. Clinical & Experimental Metastasis. 2018;35(3):123–134. doi: 10.1007/s10585-018-9894-x. [DOI] [PubMed] [Google Scholar]
- 223.Yin C., Evason K. J., Asahina K., Stainier D. Y. R. Hepatic stellate cells in liver development, regeneration, and cancer. Journal of Clinical Investigation. 2013;123(5):1902–1910. doi: 10.1172/JCI66369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Grünwald B., Harant V., Schaten S., et al. Pancreatic premalignant lesions secrete tissue inhibitor of metalloproteinases-1, which activates hepatic stellate cells via CD63 signaling to create a premetastatic niche in the liver. Gastroenterology. 2016;151(5):1011–1024.e7. doi: 10.1053/j.gastro.2016.07.043. [DOI] [PubMed] [Google Scholar]
- 225.Fernandez M. C., Rayes R., Ham B., et al. The type I insulin-like growth factor regulates the liver stromal response to metastatic colon carcinoma cells. Oncotarget. 2017;8(32):52281–52293. doi: 10.18632/oncotarget.12595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Piquet L., Dewit L., Schoonjans N., et al. Synergic interactions between hepatic stellate cells and uveal melanoma in metastatic growth. Cancers. 2019;11(8):p. 1043. doi: 10.3390/cancers11081043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Babchia N., Landreville S., Clément B., Coulouarn C., Mouriaux F. The bidirectional crosstalk between metastatic uveal melanoma cells and hepatic stellate cells engenders an inflammatory microenvironment. Experimental Eye Research. 2019;181:213–222. doi: 10.1016/j.exer.2019.02.012. [DOI] [PubMed] [Google Scholar]
- 228.Heldin C. H., Rubin K., Pietras K., Östman A. High interstitial fluid pressure -- an obstacle in cancer therapy. Nature Reviews Cancer. 2004;4(10):806–813. doi: 10.1038/nrc1456. [DOI] [PubMed] [Google Scholar]
- 229.Mazza G., al-Akkad W., Rombouts K. Engineering _in vitro_ models of hepatofibrogenesis. Advanced Drug Delivery Reviews. 2017;121:147–157. doi: 10.1016/j.addr.2017.05.018. [DOI] [PubMed] [Google Scholar]
- 230.Ouchi R., Togo S., Kimura M., et al. Modeling steatohepatitis in humans with pluripotent stem cell-derived organoids. Cell Metabolism. 2019;30(2):374–384.e6. doi: 10.1016/j.cmet.2019.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Fugaru I., Bérubé J., Babchia N., Coulouarn C., Mouriaux F., Landreville S. Uncovering the mechanisms behind hepatic microenvironment remodeling in metastatic uveal melanoma. Investigative Ophthalmology & Visual Science. 2017;58(8):p. 3968. [Google Scholar]
- 232.Sun W., Luo Z., Lee J., et al. Organ-on-a-chip for cancer and immune organs modeling. Advanced Healthcare Materials. 2019;8 doi: 10.1002/adhm.201801363. [DOI] [PubMed] [Google Scholar]
- 233.Mosaad E., Chambers K., Futrega K., Clements J., Doran M. R. Using high throughput microtissue culture to study the difference in prostate cancer cell behavior and drug response in 2D and 3D co-cultures. BMC Cancer. 2018;18(1):p. 592. doi: 10.1186/s12885-018-4473-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.O'Connor K. C. Three-dimensional cultures of prostatic cells: tissue models for the development of novel anti-cancer therapies. Pharmaceutical Research. 1999;16(4):486–493. doi: 10.1023/A:1011906709680. [DOI] [PubMed] [Google Scholar]
- 235.Namekawa T., Ikeda K., Horie-Inoue K., Inoue S. Application of prostate cancer models for preclinical study: advantages and limitations of cell lines, patient-derived xenografts, and three-dimensional culture of patient-derived cells. Cells. 2019;8(1):p. 74. doi: 10.3390/cells8010074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Ding Y., Liu W., Yu W., et al. Three-dimensional tissue culture model of human breast cancer for the evaluation of multidrug resistance. Journal of Tissue Engineering and Regenerative Medicine. 2018;12(9):1959–1971. doi: 10.1002/term.2729. [DOI] [PubMed] [Google Scholar]
- 237.Hume R. D., Berry L., Reichelt S., et al. An engineered human adipose/collagen model for in vitro breast cancer cell migration studies. Tissue Engineering Part A. 2018;24(17-18):1309–1319. doi: 10.1089/ten.TEA.2017.0509. [DOI] [PubMed] [Google Scholar]
- 238.Delort L., Lequeux C., Dubois V., et al. Reciprocal interactions between breast tumor and its adipose microenvironment based on a 3D adipose equivalent model. PLoS One. 2013;8(6, e66284) doi: 10.1371/journal.pone.0066284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Belgodere J. A., King C. T., Bursavich J. B., Burow M. E., Martin E. C., Jung J. P. Engineering breast cancer microenvironments and 3D bioprinting. Frontiers in Bioengineering and Biotechnology. 2018;6:p. 66. doi: 10.3389/fbioe.2018.00066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Guiro K., Arinzeh T. L. Bioengineering models for breast cancer research. Breast Cancer: Basic and Clinical Research. 2015;9(Supplement 2):57–70. doi: 10.4137/BCBCR.S29424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Ellem S. J., de-Juan-Pardo E. M., Risbridger G. P. In vitro modeling of the prostate cancer microenvironment. Advanced Drug Delivery Reviews. 2014;79-80:214–221. doi: 10.1016/j.addr.2014.04.008. [DOI] [PubMed] [Google Scholar]
- 242.Aponte P. M., Caicedo A. Stemness in cancer: stem cells, cancer stem cells, and their microenvironment. Stem Cells International. 2017;2017:17. doi: 10.1155/2017/5619472.5619472 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Beaudet M. J., Yang Q., Cadau S., et al. High yield extraction of pure spinal motor neurons, astrocytes and microglia from single embryo and adult mouse spinal cord. Scientific Reports. 2015;5(1) doi: 10.1038/srep16763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Chabaud S., Marcoux T. L., Deschênes-Rompré M. P., et al. Lysophosphatidic acid enhances collagen deposition and matrix thickening in engineered tissue. Journal of Tissue Engineering and Regenerative Medicine. 2015;9(11):E65–E75. doi: 10.1002/term.1711. [DOI] [PubMed] [Google Scholar]
- 245.Ayoub A., Pereira J. M., Rioux L. E., Turgeon S. L., Beaulieu M., Moulin V. J. Role of seaweed laminaran from Saccharina longicruris on matrix deposition during dermal tissue-engineered production. International Journal of Biological Macromolecules. 2015;75:13–20. doi: 10.1016/j.ijbiomac.2015.01.017. [DOI] [PubMed] [Google Scholar]
- 246.Santoro M., Lamhamedi-Cherradi S. E., Menegaz B. A., Ludwig J. A., Mikos A. G. Flow perfusion effects on three-dimensional culture and drug sensitivity of Ewing sarcoma. Proceedings of the National Academy of Sciences. 2015;112(33):10304–10309. doi: 10.1073/pnas.1506684112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Bruce A., Evans R., Mezan R., et al. Three-dimensional microfluidic tri-culture model of the bone marrow microenvironment for study of acute lymphoblastic leukemia. PLoS One. 2015;10(10, e0140506) doi: 10.1371/journal.pone.0140506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Ferrarini M., Steimberg N., Boniotti J., et al. 3D-dynamic culture models of multiple myeloma. Methods in Molecular Biology. 2017;1612:177–190. doi: 10.1007/978-1-4939-7021-6_13. [DOI] [PubMed] [Google Scholar]
- 249.Trachtenberg J. E., Santoro M., Williams C., III, et al. Effects of shear stress gradients on ewing sarcoma cells using 3D printed scaffolds and flow perfusion. ACS Biomaterials Science & Engineering. 2017;4(2):347–356. doi: 10.1021/acsbiomaterials.6b00641. [DOI] [PubMed] [Google Scholar]
- 250.Di Sanzo M., Cipolloni L., Borro M., et al. Clinical applications of personalized medicine: a new paradigm and challenge. Current Pharmaceutical Biotechnology. 2017;18(3):194–203. doi: 10.2174/1389201018666170224105600. [DOI] [PubMed] [Google Scholar]
- 251.Collins F. S., Varmus H. A new initiative on precision medicine. New England Journal of Medicine. 2015;372(9):793–795. doi: 10.1056/NEJMp1500523. [DOI] [PMC free article] [PubMed] [Google Scholar]