Abstract
Tumor-stroma interactions have emerged as critical determinants of drug efficacy. However, the underlying biological and physicochemical mechanisms by which the microenvironment regulates therapeutic response remain unclear, due in part to a lack of physiologically relevant in vitro platforms to accurately interrogate tissue-level phenomena. Tissue-engineered tumor models are beginning to address this shortcoming. By allowing selective incorporation of microenvironmental complexity, these platforms afford unique access to tumor-associated signaling and transport dynamics. This review will focus on engineering approaches to study drug delivery as a function of tumor-associated changes of the vasculature and extracellular matrix (ECM). First, we review current biological understanding of these components and discuss their impact on transport processes. Then, we evaluate existing microfluidic, tissue engineering, and materials science strategies to recapitulate vascular and ECM characteristics of tumors, and finish by outlining challenges and future directions of the field that may ultimately improve anti-cancer therapies.
1. Introduction
Given its extensive socioeconomic impact, cancer continues to be a major focus of drug development and delivery research. Nevertheless, clinical success of anti-cancer therapies remains limited, and most treatment strategies exhibit marginal efficacy, serious side effects, and the development of resistance. Moreover, complete tumor eradication is mostly impossible, and time until patient relapse or metastasis remains a tragic measure of clinical success. Targeted therapies interfering with specific genetic and molecular mechanisms of tumorigenesis have offered improvement relative to conventional cytotoxic therapy; however, cancer cells frequently evade therapy by assuming resistance mechanisms including secondary mutations and epigenetic modifications [1-3].
While many therapies directly target tumor cells, the microenvironment in which tumor cells reside is an equally important participant in disease progression. During health, normal “contextual cues” of the host microenvironment prevent the cancerous outgrowth of epithelial cells [4, 5]. However, perturbation of this homeostasis, e.g., due to chronic inflammation, metabolic changes, or hormonal imbalance, enables the initiation and progression of malignancy [6-9] as well as the emergence of resistance [10, 11].
In addition to directly affecting tumor cell behavior, microenvironmental conditions may promote recurrence by simply preventing effective transport of therapeutics. When anti-cancer drugs are systemically administered, steps of drug delivery include transport (1) within the circulation, (2) across blood vessel walls, and (3) through the interstitial space to the tumor [12, 13]. Alterations of microenvironmental conditions interfering with any of these processes may affect drug bioavailability with consequences on efficacy.
The physicochemical properties of the vasculature and the interstitial extracellular matrix (ECM) are key regulators of anti-cancer drug distribution and efficacy [14]. As the primary conduits of perfusion, blood vessels determine the availability of drugs throughout the body and within individual tissues. However, heterogeneous microvascular function as present within tumors can compromise delivery and undermine the effects of therapeutic agents [14]. Enhanced permeability and retention (EPR) in leaky vessels has facilitated the targeting of macromolecular therapies [15-19]. Yet, the asymmetric distributions of oxygen or drugs within a tumor provide a conducive landscape for the evolution of resistance within heterogeneous populations of cancer cells [20]. Although vascular structure and function largely regulate the spatiotemporal distribution of drug, interstitial space can also affect transport rates [21]. In particular, excessive ECM deposition due to fibrotic remodeling (also termed desmoplasia) physically hinders diffusion of large anti-tumor molecules through the interstitium [21].
Despite the well-established physical principles governing biological transport phenomena, the opportunity to leverage these principles to improve therapeutic outcomes is limited. Conventionally, new anti-cancer compounds are first tested in 2D tissue culture, which provide homogeneous access to drug and neglect the 3D microenvironmental properties inherent to tumors. Additionally, even positive results from animal studies do not always translate to efficacy in humans due to species-dependent discrepancies in signaling and physiology [22, 23]. The development of tissue-engineered model systems that accurately recapitulate human tumor with increasing physiological complexity may help to understand and test microenvironmental parameters affecting tumor response. Here, we review current understanding of the biological characteristics underlying tumor-associated changes of the vasculature and ECM properties, examine the consequences of these parameters for mass transport and drug delivery, and present emerging in vitro strategies that may provide new insights for improved anti-cancer therapies [24-26].
2. Tumor vasculature: biophysical changes and their relevance to drug delivery
2.1) Biological characteristics of tumor microvasculature
Since Judah Folkman’s seminal observations in 1971 that tumorigenesis is associated with the ingrowth of abnormal blood vessels [27, 28], vascular dysfunction has become an enduring theme in cancer biology and anti-cancer therapy. When compared to healthy vasculature, tumor vessels are leaky, fenestrated, tortuous, and dilated, with chaotic branching patterns including shunts and loops, as well as irregular hierarchy of vessels [29-34] (Figure 1). The tumor vasculature comprises at least six types of vascular structures with distinct properties, including mother vessels (MVs) and several varieties of daughter vessels (capillaries, glomeruloid microvascular proliferations, vascular malformations) [35, 36]. MVs are enlarged sinusoids resulting from pericyte detachment, basal membrane (BM) degradation, and endothelial thinning [36]. Despite this distension, MVs do not increase blood flow, possibly due to their hyper-permeable membrane function [35]. Meanwhile, immature daughter vessels are fenestrated and lack functional perfusion. Vascular leakage, coupled to dysfunctional lymphatic drainage, results in an accumulation of interstitial fluid, which compromises the hydrostatic and osmotic pressure gradients that drive transvascular convection [37]. Collectively, this heterogeneous network of aberrant blood vessels yields erratic perfusion of diseased tissue, with important consequences for pathogenesis and therapy.
Figure 1. Biophysical changes of tumor vasculature.
A) Schematic illustration of the structural and functional changes in tumor vasculature, including tortuous vessels, chaotic branching, disrupted barrier function, and compromised hydrostatic and osmotic gradients. B) Aberrant vasculature undermines transport processes within diseased tissue, resulting in asymmetric distributions of oxygen and drugs to the tumor. This micrograph illustrates the penetration depth of doxorubicin and highlights the large, hypoxic regions of the tumor unaffected by therapy [48]. Reproduced with permission from Nature Publishing Group.
Hypoxia and acidosis are the most prominent consequences of deregulated vascular function. Compromised vessel characteristics lead to poor supply of oxygen and clearance of metabolic wastes, mediating sustained hypoxia (<1% O2) and acidosis (as low as pH ~6.6-6.8 in some areas) [38-40]. These microenvironmental conditions directly promote tumor progression and new vessel formation by activating a variety of transcriptional programs [41-44]. In particular, stabilization of the alpha subunit of the hypoxia inducible factor 1 (HIF-1) transcription factor in low-oxygen conditions leads to an orchestrated program of hypoxic response [42, 45-47] that includes the up-regulation of pro-angiogenic morphogens (including vascular endothelial growth factor [VEGF] and basic fibroblast growth factor [bFGF]). However, rather than promoting the formation of healthy blood vessels that could normalize tissue O2 levels and pH, excess proangiogenic signaling activates a vicious cycle that undermines vascular stability by impairing vessel organization and permeability thus exacerbating these pathological conditions [48-50].
In addition to metabolic transport, impaired vascular function also compromises the homogeneous delivery of therapeutic agents, resulting in poor distribution of drugs throughout the tumor [51]. Low vascular function at the tumor interior prevents therapeutic access to large regions of tissue [31]. Homogeneous delivery is further undermined by the absence of hydrostatic and osmotic pressure gradients, which are necessary for interstitial convection to distribute large therapeutic agents [52, 53]. In some cases, enhanced vascular permeability and retention (EPR) has been coopted as a mechanism for tumor targeting of large particles such as antibodies and micelles [15]. However, although high molecular weight drugs and drug carriers easily traverse the endothelial membrane of the tumor, they have poor penetration depth and achieve little benefit in regions distal to blood vessels [51, 54, 55]. Therefore, vascular normalization is an emerging theme for improving therapeutic delivery [56-58].
Whereas most vascular therapies emphasize the destruction of blood vessels to stunt tumor growth, some researchers hope to re-appropriate angiogenic drugs as adjuvant therapy in order to improve drug distribution [59]. By increasing perfusion in the tumor, this method may overcome major disadvantages of chemotherapy such as short half-life and small therapeutic index (range of concentration between efficacy and toxicity) [60]. In the past decade, numerous clinical trials have revealed the benefits of anti-angiogenic therapy as an adjuvant to chemotherapy or radiation therapy [60-62]. However, the dearth of pre-clinical models to recapitulate vascular transport remains a challenge for the development of new strategies for vascular normalization.
2.2) Effects of microvascular dysfunction on transport physics
Whereas conventional pharmacokinetic metrics (e.g. distribution volume, half-life, clearance rate) characterize the average availability of drug in tissues, these measurements only poorly approximate the effective distribution of agents within tumors. In fact, large regions of tumors lack adequate vascular perfusion and thus, can remain unaffected by treatment, even at high doses. This is particularly true of large molecules (>10 kDa), which are not able to freely diffuse across the endothelium and through the tissue. Instead, convective forces govern the exchange of large compounds from the circulation (Box 1). In healthy tissues, these forces balance the influx and outflux at the arterioles and post-capillary venules to provide adequate perfusion. In the case of a tumor, however, vascular leakiness reduces hydrostatic pressure in the vessel while increasing osmotic pressure in the interstitium (approximately 29 +/− 3 mmHg in breast tumor, compared to −0.3 +/− 0.1 mmHg in healthy breast tissue) [63, 64]. Together, these changes significantly diminish the bioavailability of drugs by reducing transcapillary hydrostatic gradients necessary for fluid exchange [65].
Box 1. Forces governing transvascular transport.
The Kedem-Katchalsky equation describes solute flux(Js) as a combination of convective and diffusive forces across the endothelial membreane:
Where σf is the filtration reflection coefficient, is the average molar concentration in the membrane, P is the permeability cofficient, S is the surface area, and ΔC is the change in concentration across the endothelium. The fluid flux (Jv) is derived from Staling’s Law, which describes the contributions of hydrostatic (p) and osmotic (π) pressure gradients:
Where Lp is hydraulic conductivity, S is surface area, and σs is the osmotic reflection cofficient, which ranges from 0–1 based on the membrane permeability to solutes. Based on the equations, tranvascular solute transport is governed by four physiological constants describing solute and membrane characteristics:
Measurements for these parameters in healthy and tumor tissue are provided by [53]. These factors are affected by solute (size, charge, polarity) and membrane (pore size, pore density) properties.
In addition to simple mass transport equations, a variety of tools have been developed to quantitatively evaluate the effectiveness of mass transport in tissues. Dimensionless numbers are particularly useful to identify transport limitations [66]. For example, the Biot number compares the relative resistance attributed to the endothelial membrane versus the interstitial matrix. Likewise, the effectiveness factor describes the rate of transport compared to the rate of reaction. Both of these parameters can help identify limitations to drug penetration and efficacy. For example, in cases where transvascular transport limits therapeutic efficiency, combinations with permeabilizing agents may improve delivery, while other therapies limited by interstitial diffusion may benefit from stroma-targeted treatments. A modified effectiveness factor can incorporate the Biot number to identify the relative resistance due to vascular versus interstitial transport [67, 68].
Similarly, the “observable modulus” evaluates penetration distance based on transport and reaction considerations [67]. This parameter defines a “dimensionless reaction rate” (the effectiveness factor * thiele modulus), and is defined as a function of the distance from the nearest vessel. This parameter allows the calculation of a penetration depth, or the distance at which the concentration drops below an effective dose. A low modulus indicates deeper penetration, and high modulus indicates poor distribution away from the capillary. The observable modulus has been used to determine the Krogh distance, or the range for diffusion-limited oxygen transport before reaching anoxia and necrosis [67]. In the case of therapy, the same parameter can describe the effective therapeutic range of a drug and thus characterize and inform angiogenic therapy or EPR designs.
By evaluating the specific physical contributions to therapeutic resistance by transport or reaction kinetics, these quantitative methods may help identify major barriers of delivery and enable the rational design of dosing strategies, including in combination with drugs that target the vasculature or the stroma to optimize therapeutic efficacy. Nevertheless, many of these quantitative approaches are frequently neglected. This may be due in part to the inaccessibility of the various coefficients needed to determine the dimensionless parameters. Conventional methods to test vascular permeability, such as Evan’s blue staining, provide little chance for rigorous quantification. Meanwhile, in vitro methods to measure endothelial permeability or tissue diffusivity, such as multilayer or spheroid cultures, may not accurately reflect in vivo parameters [55]. Therefore, new methods to accurately determine the parameters affecting vascular and interstitial mass transport will provide a valuable resource for the design of therapeutic strategies that target the tumor microenvironment [66].
2.3) Recapitulating tumor-associated vascular transport through in vitro models
Organotypic 3D culture models present an emerging opportunity to create controlled experimental conditions that faithfully reproduce aspects of the physiological environment. However, as sufficient mass transport within cell-laden biomaterials remains a significant limitation of 3D cultures, these approaches are typically performed in a microscale format and lack the recapitulation of tumor-inherent transport physics. New in vitro platforms that integrate functional vascular structures can help to overcome this limitation [69]. Biomimetic microvascular networks provide controlled experimental platforms to evaluate specific interactions between endothelial cells and tumor environment, improve the design of therapies targeting the vasculature, and assess the efficacy of new drugs or drug delivery strategies [70, 71].
Two categories of microfluidic vascular models have been introduced. The first design, pioneered by the groups of Roger Kamm and Lance Munn, employs microfabricated silicone molds to confine biological hydrogels between parallel microfluidic channels (Figure 2A). Endothelial cells cultured on the surface of these confined gels are able to sprout into the matrix and recapitulate aspects of angiogenic invasion, including sensitivity to shear stress, interstitial flow, and various angiogenic gradients [72-74]. These assays have been used to facilitate co-culture with stromal or tumor cells and evaluate paracrine effects in angiogenesis [74]. However, these models are confined by thin geometries (~100 μm) and are therefore sensitive to boundary effects due to cell and molecular interactions with the device surface. In addition, the endothelial cells initially form a monolayer against the gel region, rather than a fully-mimetic lumen structure. Therefore, more advanced models of microvascular structure and function have recently been developed.
Figure 2. Examples of microengineered vascular structures.
A) Vascular structures created by culturing endothelial cells in confined gels within a microfluidic platform are useful for evaluating endothelial cell response to angiogenic gradients, shear stresses, or paracrine interactions [64]. B) Anastomosis between parallel vessels enables the autonomous formation of capillary-scale networks fully embedded within 3D biological matrices. The original vessel structures were fabricated by the pull-through method [66]. C) Live/dead staining illustrates dependence of cell viability on spatial differences in oxygen and nutrient supply caused by diffusion-limited transport from perfused vascular channels. Vessels were produced by molding a hydrogel against a bioprinted sacrificial scaffold [68]. D) Vascular networks enabling control over endothelial cell (CD31) interactions with pericytes (α-SMA) and membrane permeability (fluorescein leakage) permit complex studies of vascular biology in healthy and disease conditions. Networks were generated in hydrogels by soft lithography [72]. Images were reproduced with permission from the Royal Society of Chemistry (A) and the National Academy of Sciences, USA (B-D).
This second type of platform comprises bona fide vascular structures fully embedded within 3D ECM. Such structures can be developed by molding biomaterials against a thin needle to define a vessel lumen and subsequently seeded with endothelial cells [75, 76] (Figure 2B). These vessels allow high throughput and facile fabrication, but they are restricted to simple linear geometries and are limited by the resolution of the needle. More complex channel configurations can be fabricated by patterning of a sacrificial resist that dissolves after gel crosslinking [77, 78] (Figure 2C), or by replica molding and soft lithography methods, originally developed by George Whitesides group in silicone plastics [79, 80]. Recently, advances in this technique allowed patterning of cell-laden biological materials [81, 82] and the perfusion of scaffolds with endothelialized microvascular channels [77, 78, 82, 83] (Figure 2D). Although lithographic techniques are limited to planar configurations, the bioprinted sacrificial molds were capable of generating a lattice of interconnected vessels [78]. Finally, modular assembly of cell-laden beads permits the formation of contiguous endothelial architectures, although the geometries of these networks are less defined than the other approaches [84, 85]. Together, these platforms enable unprecedented access to questions in vascular biology and vascularized tissue function [86].
In vitro microvascular platforms are particularly suited to quantify physiological parameters governing mass transport and drug delivery [76, 82]. For example, microfabricated vascular structures permit measuring endothelial permeability in authentic lumenized channels as a function of pericyte coverage, and in response to biochemical factors known to interfere with endothelial cell signaling [82]. Furthermore, microvascular platforms offer exquisite opportunity to measure cell viability as a function of Krogh distance and metabolic consumption at specific cell densities [78, 81] and to monitor transvascular and interstitial transport of therapeutic agents. Finally, they provide increasingly accurate models of the tumor environment, allowing tumor-stromal cell cross talk in the assessment of efficacy [74, 87-90]. Taken together, engineering models of vascular transport facilitate pre-clinical studies of drug delivery with human cells within a biomimetic tissue environment.
3. Tumor desmoplasia and ECM remodeling: biophysical changes and their relevance to drug delivery
3.1) Biology of tumor desmoplasia and ECM remodeling
As tumors progress from a benign to malignant stage, they recruit cancer-associated fibroblasts (CAFs) that can modulate drug response through altering ECM physicochemical properties (Figure 3). CAFs, which include myofibroblasts, deposit abundant amounts of fibrillar ECM molecules including collagen I and fibronectin [91]. These ECM compositional changes entail structural and mechanical alterations of the ECM [92-94]. For example, CAFs mediate the partial unfolding of fibronectin, which increases both the stiffness of individual fibronectin fibers [95, 96] and their ability to bind other ECM molecules such as glycosaminoglycans (GAGs) and collagen [97]. Along with elevated collagen crosslinking (e.g., by lysyl oxidase, non-enzymatic glycation, or transglutamination [98, 99]) and GAG concentration [93], these pronounced changes in ECM density and intermolecular interactions globally enhance tumor stiffness with direct consequences for tumor progression.
Figure 3. Biophysical changes of mammary tumor stroma.
A) Schematic illustration of the structural and functional changes in tumor-associated mammary stroma. Tumor-derived cytokines not only promote recruitment of inflammatory cells, but also cancer-activated fibroblasts, which stiffen tumors by thickening, linearizing and aligning fibrillar ECM components. These changes alter the transport of growth factors and cytokines, but also of therapeutic compounds with consequences on cancer cell drug resistance. B) Visualization of collagen structure in mammary tissue of normal and tumor-bearing mice via second harmonic generation imaging. Coiled and disorganized collagen fibers are present in normal mammary tissue. Upon tumorigenesis, thicker and linearized collagen fibers develop, which are tangentially and radially oriented along the boundary of premalignant and invasive mammary tumors, respectively [121, 122]. Reproduced with permission from Biomed Central.
Despite this overall increase in matrix deposition and cross-linking, tumor cells simultaneously mediate the degradation of the ECM, in part by secreting enzymes such as matrix metalloproteinases (MMPs). In addition to generating tumorigenic ECM fragments [100], elevated MMP activity affects the local (microscale) physical properties of the tumor ECM. For example, MMPs can govern degradation and reorganization of fibrillar collagen, while enhancing interactions with other ECM components through exposure of cryptic binding sites [101-103]. Nevertheless, it has to be kept in mind that non-proteolytic functions of MMPs may be similarly important. For example, MMP-9 has been associated with smooth muscle cell adhesion, migration, and cell-mediated collagen contraction, independent of it’s proteolytic function [103, 104]. The diverse roles of MMPs in tumorigenesis have previously been reviewed by Egeblad [105] and Page-McCaw [106].
Alterations of ECM composition, structure, and mechanics stimulate malignancy and drug resistance by inducing manifold changes in cell signaling [107-110]. In general, cell-ECM interactions facilitate the clustering of integrin adhesion receptors leading to cytoskeletal reorganization and cell contractility [108, 111]. These changes activate signaling cascades that play critical roles in cell fate decisions, tumor progression, and drug resistance (e.g., Rho/Rock, ERK/MAPK, and PI3K) [112-115]. For example, integrin signaling alters the co-activation of certain growth factor receptors (e.g., epidermal growth factor receptor [EGFR]) [116] which can impact targeted therapies designed to interfere with the related signaling cascades (e.g., cetuximab and panitumumab targeting EGFR, PLX4720 and PLX403 interfering with BRAF kinase activity) [117, 118]. Changes in ECM composition and exposure of cryptic binding domains partially modulate these signaling cascades by affecting integrin selectivity [119, 120]. Likewise, increased stiffness-mediated integrin signaling plays a major role in promoting cell contractility, which directly stimulates malignancy by fueling tumor cell proliferation and migration [121]. Moreover, matrix stiffness alters the behavior of tumor-associated stromal cells to drive disease progression. For example, increased stiffness promotes the pro-tumorigenic capability of mesenchymal stem cells [114, 122], compromises endothelial barrier functions [123, 124], and alters macrophage functions [125]. Finally, stiffness-dependent changes in cell contractility are critical to the myofibroblastic differentiation of stromal fibroblasts [97], thus, providing a positive feedback mechanism that further stimulates desmoplastic remodeling. For these reasons, stromal markers have the potential to serve as important indices that can help to assess and predict patient diagnostics and outcomes [126-133].
In addition to the direct mechano-biological effects, physicochemical changes in the ECM can impact soluble factor signaling [134]. Specifically, the increase of ECM components with growth factor-binding domains affects the bioavailability of these molecules. However, this process is complicated by the reciprocal interplay between multiple ECM molecules. For example, interactions with GAG (e.g. heparin) modulate fibronectin conformation and expose cryptic binding sites involved in growth factor sequestration [135]. Likewise, changes in cell contractility can expose these latent binding sites and thereby cause the release of morphogens from their ECM depots (e.g. post-translational activation of latent transforming growth factor β [TGF-β]) [136].
3.2) Effects of tumor-associated ECM on transport physics
Transport of therapeutic molecules through interstitial tissue is dependent on convection and diffusion [21]. However, the combination of leaky vasculature and dense ECM increases interstitial fluid pressure (IFP) [137, 138] and inhibits convection-mediated transport. Consequently, drug-delivery within tumor stroma primarily depends on diffusion [21]. However, dense cellular and matrix components represent diffusion barriers that hinder transport through the interstitium [139, 140]. Specific parameters that regulate diffusion efficiency through the stroma include 1) diffusion distance, 2) available volume fraction of pores (accessible space where molecules can pass through), 3) tortuosity of pathway, 4) hydrodynamic resistance, and 5) ECM affinity of the molecule of interest [67] (BOX2). All of these parameters are affected by tumor stroma remodeling. For example, desmoplasia-mediated enhancement of ECM density and structural changes decrease the available volume fraction of pores and increase the tortuosity of the void space, both of which reduce the rate of diffusion through the stroma [141, 142]. Therefore, properties of both the drug (e.g. size, charge, configuration, etc.) [143] and the ECM (e.g., composition, viscoelasticity, geometrical arrangement, and/or electrostatic properties) should be considered when approximating an agent’s bioavailability [139, 141, 144-146].
Box 2. Forces governing interstitial transport.
Solute transport through interstitial space results from the sum of convective (JC) and diffusive (JD) fluxes:
Where D is the effective diffusion coefficient, C is the solute concentration, f is the retardation coefficient, and vf is interstitial fluid velocity, determined by the solution to the Brinkman equation for flow through porous media:
Where μ is the fluid viscosity, K is the dydraulic conductivity, and p is hydrostatic pressure difference between the vascular and lymphatic vessels. In most cases, K, D, are f are approximately equivalent tot the hydraulic conductivity (Lp), permeability (P), and 1 minus the filtration reflection coefficient(1-σf) for membrane transport (see Box 1). Parameter values can be found in [12].
The specific structure of ECM components also modulates diffusion fluxes of therapeutic molecules. Analysis of collagen fiber orientation via second harmonic generation imaging (SHG) revealed that increased malignancy is associated with collagen fiber re-orientation. While collagen in initiating tumors is characterized by isotropic orientation, progression leads to tangential and ultimately radial alignment in expanded and invading tumors, respectively [147, 148] (Figure 3B). These changes in ECM fiber network orientation can promote diffusion anisotropy without affecting the overall diffusion coefficient of the drug [149]. For example, fibers tangentially aligned to the tumor boundary could redirect drug diffusion away from the tumor and, therefore, impair therapy efficacy during initial stage of tumorigenesis. Theoretically, radially aligned fibers should mediate the opposite effect; however, at this stage tumor cells may have developed resistance phenomena rendering them unresponsive to therapy.
Finally, changes in ECM composition can affect delivery by altering hydrodynamic and electrostatic interactions of the drug with the ECM. Generally, tumor-mediated elevation of ECM viscosity increases the hydrodynamic resistance of molecules and hence slows down their movement through the interstitial space [67, 139, 142, 144, 150]. These differences are aggravated by changes in ECM charge distribution (e.g. due to increased association with negatively charged sulfated GAGs including heparan sulfate [134, 144]. More specifically, electrostatic repulsion or attraction of charged particles can reduce the available volume fraction of pores and/or cause local sequestration of the drug. Furthermore, electrostatic interaction may not only be relevant to ECM-drug interactions per se, but can also alter the conformation of ECM molecules (e.g. as outlined above for fibronectin-heparin interactions [151]). These variations, in turn, can impact their ability to bind morphogens ultimately exaggerating asymmetric drug distribution within tumors.
The potential opportunity for enhancing drug transport by modifying tumor matrix was demonstrated in a mouse model of pancreatic cancer, where gemcitabine delivery was improved through depletion of desmoplastic stroma by inhibition of hedgehog signaling [152]. However, Infinity Pharmaceuticals’ phase II clinical trials for saridegib (IPI-926) in combination with gemcitabine were halted last year due to increased mortality, suggesting that this strategy may require improved understanding of tumor-stromal interactions in human patients. Consequently, tumor-associated ECM remodeling critically impacts diffusive transport, yet, most drug testing approaches neglect the contribution of the above-described parameters. Physiologically relevant 3D tumor models recapitulating the physicochemical properties of the ECM that interfere with diffusion fluxes offer promise to selectively define coefficients modulating drug transport dynamics in tumors.
3.3) In vitro models recapitulating ECM-mediated changes in tumor transport
Natural biomaterials can be employed to assess tumorigenesis as a function of altered ECM characteristics. Matrigel®, a basement membrane mixture isolated from murine Engelbreth-Holm-Swarm sarcoma, is currently most widely used. However, inherent batch-to-batch variability, incomplete controllability of physicochemical parameters (e.g. narrow range of stiffness), and lack of covalent crosslinks critical to the basement membrane barrier function [95, 153-155] represent limitations of this material. Collagen type I is a defined biological scaffold that can be synthetically modified to provide a wider range of physicochemical properties. For example, collagen stiffness can be adjusted through covalent crosslinking by non-enzymatic glycation [156]. However, this procedure can impact cell behavior by preventing enzymatic degradation of the matrix and by accumulating advanced glycation end products, both of which can affect biochemical signaling. [157].
Synthetic biomaterials have emerged as attractive alternatives to modulate stiffness in a more controlled manner. ECM-coated polyacrylamide (PA) gels allow adjustment of stiffness independent of ligand concentration. This platform has been critical to the identification of mechanoregulatory signaling mechanisms of tumorigenesis [121]. However, PA gels cannot be remodeled by cells, and the fabrication method further limits their use to 2D studies which may affect critical cell functions [110, 158]. Moreover, altering crosslinking density can affect the surface porosity of PA gels, which may cause conformational changes of the ECM coating that modulate cell behavior [159].
Poly (ethylene glycol) (PEG)-based materials can address some of these challenges. For example, remodelable PEG-based hydrogels have been developed to study the independent effects of stiffness and peptide ligand density on lung cancer cells in a 3D context (Figure 4A) [160]. Moreover, photodegradable or photoreversible crosslinks may be introduced to enable temporal control of substrate stiffness in situ [161, 162]. By allowing dynamic physicochemical changes in the ECM, such platforms can provide important new insights to tumor development. For example, studying proteolysis-mediated variations of cellular traction could reveal the temporal effects of substrate stiffness on cell fate decisions (as recently shown with hyaluronic acid-based degradable materials (Figure 4B) [163]. Furthermore, integration of growth factor binding sites could mimic the effects of matrix-binding on morphogen or drug distribution [164].
Figure 4. Examples of engineering approaches to recapitulate tumor-associated ECM characteristics.
A) Covalently cross-linked PEG-based hydrogels with controlled stiffness and ligand concentration have been used to investigate the impact of these parameters on lung cancer cells [135]. B) Tunable hyaluronic acid–based 3D hydrogels containing proteolytically cleavable crosslinks revealed a role of ECM degradation-mediated cellular traction in cell fate decisions [138] C) Studies with stretchable PDMS molds containing fibrillar collagen gels revealed that axially oriented collagen fibers promote epithelial co-orientation, which is prevented by randomly oriented collagen fibers [143]. D) Tumor-mediated changes in fibronectin conformation can be simulated with conducting polymers. Applying an appropriate potential to the cytocompatible conducting polymer poly (3,4-ethylenedioxythiophene) doped with p-toluenesulfonate (PEDOT:TOS) yields conformation gradients of adsorbed fibronectin that can be confirmed by FRET analysis and that influence cellular adhesion (green calcein staining) [126, 151]. Images were reproduced with permission from American Association for Cancer Research (A), Nature Publishing Group (B), Cell Press (C), Wiley Online Library and Royal Society of Chemistry (D).
While the above approaches rely on changing the material chemistry, ECM structural changes can also be introduced by varying the scaffold fabrication conditions. One relatively simple approach to modify collagen fibril characteristics is by adjusting the gelation temperature, pH, and/or material concentration [165]. Additionally, the collagen isolation technique is a critical consideration, as fibers from acid-solubilized collagen may mimic in vivo microarchitecture, but these gels also contain telopeptides that may compromise biocompatibility relative to pepsin-digested collagen [166, 167]. Furthermore, the random orientation of spontaneously formed collagen fibrils does not recapitulate fiber alignment in tumors. One method to orient collagen fibers is by casting collagen gels into PDMS molds and exposing them to tensile stress axially [168] (Figure 4C). However, this loading method may also modulate the behavior of the embedded cells, and interpretation of results is complicated by non-linear cellular response to combinations of mechanical forces and ECM properties [169]. Electrospinning of artificial ECM scaffolds may help to overcome some of these challenges and allow the selective manipulation of ECM structural features at relevant length scales [170-173].
Fibronectin represents an important tumor-associated ECM component whose conformation can be quantified and recapitulated using biomaterials-based approaches. FRET-based analysis as established by the Vogel lab has provided quantitative insights into tumor-mediated fibronectin unfolding [96, 174]. This unfolding impacts integrin selectivity by altering the spatial separation between the RGD adhesion and PHSRN synergy sequence. One elegant approach to study the consequences of protein conformation is to modify synthetic, otherwise nonadhesive, hydrogels with RGD and PHSRN peptides either alone or in combination [175]. Alternatively, conducting polymer substrates have been employed to unfold native fibronectin. Applying differential voltages to poly (3,4-ethylenedioxythiophene) doped with p-toluenesulfonate (PEDOT:TOS) permits selective manipulation of the conformation of adsorbed fibronectin (Figure 4D) [151]. Such systems have been used to elucidate that fibronectin conformational changes influence cell adhesion and secretory patterns for both stromal and tumor cells [176, 177]. Yet, fibronectin does not occur in isolation, but is part of complex ECM scaffolds, which can be partially mimicked with fibroblast-derived 3D ECMs. These matrices are generated by detergent-based decellularization and recapitulate various physicochemical aspects of native ECMs including fiber alignment, composition, and density [115, 178]. Furthermore, they allow the delineation of effects mediated by healthy and tumor-associated ECMs and have revealed differential cell morphology, orientation, and migration, as well as drug response in healthy versus disease conditions [179].
In order to be useful in drug discovery and screening, the in vitro platforms described above must support high-throughput testing of complex libraries of biochemical-physical cues. Recently, sophisticated high throughput methods were introduced to screen for combinational effects of ECM physicochemical parameters on cells incorporated in 3D hydrogels [180]. These high throughput microarrays, combined with topographically-controllable biomaterial-based scaffolds, afford a more comprehensive understanding of the integrated effects of the tumor microenvironment on drug delivery and efficacy.
4. Future directions
Artificial cell culture platforms have traditionally benefited from the ability to isolate individual biological parameters in a controlled experimental environment, but many challenges exist when moving toward increasingly realistic models for replicating tumor pathology and therapy. Specifically, such biomimetic cultures are confined by boundaries of space, time, and complexity. Addressing these limitations will help to accurately study cancer biology and translate the findings to clinical practice.
Spatial resolution is a critical parameter of engineered tissues. Although microfabrication technologies afford unprecedented access to cell-scale precision, integrating this resolution into meaningful tissue or organ-level organization is nontrivial. For example, artificial tumors grown in confined structures may undergo differential morphogenesis relative to human disease. In addition, most microfluidic geometries present boundary artifacts, including mechanical and chemical interactions along the walls of the device that may contribute to this end [181-183]. Moreover, engineered tumors are often isolated systems that lack an organism-level context. Recent integration of multiple organ compartments into body-on-a-chip devices has started to simulate aspects of organism physiology, including drug metabolism and toxicity of anti-cancer drugs [184, 185]. These in vitro models provide a first glimpse at holistic consequences of tumor growth or therapy; however, they still neglect critical physiological characteristics, including cellular transport through the blood contributing to tumor progression or metastatic homing. For example, the molecular communication between breast cancer cells and bone marrow-derived progenitor cells may contribute to premetastatic niche formation in the target organ [186]. Systemic in vitro models that connect diverse organ compartments through engineered vascular systems may facilitate a more comprehensive understanding of the mechanisms underlying metastasis and define future treatment strategies [187, 188].
The temporal scale of in vitro models introduces a second experimental obstacle. Culture conditions of days to weeks must simulate emergent phenomena of months and years. This challenge is especially relevant in the evolution of drug resistance, where population drift relies on many generations of cell division [189, 190]. In vitro platforms may approximate these effects of tumor heterogeneity by cell sourcing from primary tumors or by seeding mixed cell populations representing different stages of tumor development [191]. In addition to drug resistance, morphological changes in tissue stroma, including vascular and ECM remodeling, often require longer durations than conventional experimental techniques. This motivates the use of artificial factors such as phorbol 12-myristate 13-acetate (PMA) to rapidly stimulate cellular response [192, 193], or the use of low density matrices to accelerate remodeling and migration [194, 195]. Although useful experimental tools, these artifices may compromise the integrity of the in vitro system. Computational models of tumorigenesis offer an alternative strategy for recapitulating the time-scale of pathogenesis [196]. For example, multiscale computational models of the tumor microenvironment can be used to predict changes in tumor malignancy as a function of stroma-dependent selective pressure [20]. However, in order to mimic physiological growth rates, these models rely on the accurate measurement of cell kinetics. By providing controlled platforms to measure these critical coefficients, in vitro tumor models may inform the development of accurate in silico models of progression and response.
Physiological complexity, including matrix composition, cellular components, or spatiotemporal presentation of soluble factors, introduces additional compromises for in vitro tissue culture. Notably, increasing complexity often undermines the controllability of the system. For example, although in vitro tumor models enable human-specific drug screening, tumor cells have a broad spectrum of characteristics depending on originating organ, stage or malignancy, and patients’ genetic background, additional health complications, and life style. Therefore, short-term culture of cell lines may not cover organ-, patient-, and stage-specific features of tumors. Furthermore, genomic and epigenomic instability leads to diverse clonal populations within a single tumor that cross-communicate [197, 198], and this signaling is lost given the uniformity of conventional cell culture lines. Advances in personalized medicine have begun to appreciate the unique molecular signature of individual patients. Integration of patient-derived biopsies within engineered models may allow personalized drug screening prior to treatment. The advantages of closely approximating physiological complexity is an asset of engineered tumor models, but remembering these limitations will help guide the interpretation and translation of data collected with these systems.
5. Conclusion
By integrating cancer biology and engineering approaches, realistic tumor models may be developed that have the potential to advance current anti-cancer therapies. More specifically, this approach will help to better define qualitative and quantitative parameters currently complicating drug transport in tumor-associated blood vessels and stroma. In the future, however, further improvements are needed to enhance the relevance of these models. For example, integration with organ-on-a-chip devices may not only help to recapitulate systemic aspects of the disease, but also to better define the spatiotemporal relationships and patient-inherent complexity complicating drug transport in the clinic. Together with current advances in cancer (epi)-genomics and computational modeling engineered tumor models offer promise to bridge the gap between bench to bedside research and help translate new therapeutic approaches to the clinic.
6. Acknowledgements
Funding was provided by a NSF graduate research fellowship for P. DelNero, the Clinical and Translational Science Center at Weill Cornell Medical College as well as the National Cancer Institute (R21CA161532, R01CA173083, and by the Cornell Center on the Microenvironment & Metastasis through Award Number U54CA143876). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
7. References
- [1].Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, Nickerson E, Auclair D, Li L, Place C, Dicara D, Ramos AH, Lawrence MS, Cibulskis K, Sivachenko A, Voet D, Saksena G, Stransky N, Onofrio RC, Winckler W, Ardlie K, Wagle N, Wargo J, Chong K, Morton DL, Stemke-Hale K, Chen G, Noble M, Meyerson M, Ladbury JE, Davies MA, Gershenwald JE, Wagner SN, Hoon DS, Schadendorf D, Lander ES, Gabriel SB, Getz G, Garraway LA, Chin L. A landscape of driver mutations in melanoma. Cell. 2012;150:251–263. doi: 10.1016/j.cell.2012.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Paraiso KH, Fedorenko IV, Cantini LP, Munko AC, Hall M, Sondak VK, Messina JL, Flaherty KT, Smalley KS. Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. British journal of cancer. 2010;102:1724–1730. doi: 10.1038/sj.bjc.6605714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Chin L, Andersen JN, Futreal PA. Cancer genomics: from discovery science to personalized medicine. Nature medicine. 2011;17:297–303. doi: 10.1038/nm.2323. [DOI] [PubMed] [Google Scholar]
- [4].Deng Y.L. Guoren, Zlotnikov Galina, Thor Ann D., Smith Helene S. Loss of Heterozygosity in Normal Tissue Adjacent to Breast Carcinomas. Science. 1996;274:2057–2059. doi: 10.1126/science.274.5295.2057. [DOI] [PubMed] [Google Scholar]
- [5].Bissell MJ, Radisky D. Putting tumours in context. Nat. Rev. Cancer. 2001;1:46–54. doi: 10.1038/35094059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Caino MC, Chae YC, Vaira V, Ferrero S, Nosotti M, Martin NM, Weeraratna A, O’Connell M, Jernigan D, Fatatis A, Languino LR, Bosari S, Altieri DC. Metabolic stress regulates cytoskeletal dynamics and metastasis of cancer cells. The Journal of clinical investigation. 2013;123:2907–2920. doi: 10.1172/JCI67841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
- [8].Park J, Euhus DM, Scherer PE. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocrine reviews. 2011;32:550–570. doi: 10.1210/er.2010-0030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nature reviews. Cancer. 2011;11:886–895. doi: 10.1038/nrc3174. [DOI] [PubMed] [Google Scholar]
- [10].Straussman T.M. Ravid, Shee Kevin, Barzily-Rokni Michal, Qian Zhi Rong, Du Jinyan, Davis Ashli, Mongare Margaret M., Gould Joshua, Frederick Dennie T., Cooper Zachary A., Chapman Paul B., Solit David B., Ribas Antoni, Lo Roger S., Flaherty Keith T., Ogino Shuji, Wargo Jennifer A., Golub Todd R. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature Letter. 2012;487:500–504. doi: 10.1038/nature11183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].McMillin DW, Negri JM, Mitsiades CS. The role of tumour-stromal interactions in modifying drug response: challenges and opportunities. Nature reviews. Drug discovery. 2013;12:217–228. doi: 10.1038/nrd3870. [DOI] [PubMed] [Google Scholar]
- [12].Jain RK. Transport of molecules in the tumor interstitium- a review. Cancer Research. 1987;47:3039–3051. [PubMed] [Google Scholar]
- [13].Jain RK. Transport of molecules, particles, and cells in solid tumors. Annual review of biomedical engineering. 1999;1:241–263. doi: 10.1146/annurev.bioeng.1.1.241. [DOI] [PubMed] [Google Scholar]
- [14].Jain RK. Barriers to drug-delivery in solid tumors. Scientific American. 1994;271:58–65. doi: 10.1038/scientificamerican0794-58. [DOI] [PubMed] [Google Scholar]
- [15].Iyer AK, Khaled G, Fang J, Maeda H. Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today. 2006;11:812–818. doi: 10.1016/j.drudis.2006.07.005. [DOI] [PubMed] [Google Scholar]
- [16].Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release. 2000;65:271–284. doi: 10.1016/s0168-3659(99)00248-5. [DOI] [PubMed] [Google Scholar]
- [17].Fang J, Nakamura H, Maeda H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced drug delivery reviews. 2011;63:136–151. doi: 10.1016/j.addr.2010.04.009. [DOI] [PubMed] [Google Scholar]
- [18].Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Advanced drug delivery reviews. 2011;63:131–135. doi: 10.1016/j.addr.2010.03.011. [DOI] [PubMed] [Google Scholar]
- [19].Davis ME, Chen Z, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nature Reviews Drug Discovery. 2008;7:771–782. doi: 10.1038/nrd2614. [DOI] [PubMed] [Google Scholar]
- [20].Anderson AR, Weaver AM, Cummings PT, Quaranta V. Tumor morphology and phenotypic evolution driven by selective pressure from the microenvironment. Cell. 2006;127:905–915. doi: 10.1016/j.cell.2006.09.042. [DOI] [PubMed] [Google Scholar]
- [21].Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nature Reviews Clinical Oncology. 2010;7:653–664. doi: 10.1038/nrclinonc.2010.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Singh M, Ferrara N. Modeling and predicting clinical efficacy for drugs targeting the tumor milieu. Nature biotechnology. 2012;30:648–657. doi: 10.1038/nbt.2286. [DOI] [PubMed] [Google Scholar]
- [23].Richert L, Tuschl G, Viollon-Abadie C, Blanchard N, Bonet A, Heyd B, Halkic N, Wimmer E, Dolgos H, Mueller SO. Species differences in the response of liver drug-metabolizing enzymes to (S)-4-O-tolylsulfanyl-2-(4-trifluormethyl-phenoxy)-butyric acid (EMD 392949) in vivo and in vitro. Drug metabolism and disposition: the biological fate of chemicals. 2008;36:702–714. doi: 10.1124/dmd.107.018358. [DOI] [PubMed] [Google Scholar]
- [24].Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. Nature reviews. Cancer. 2004;4:839–849. doi: 10.1038/nrc1477. [DOI] [PubMed] [Google Scholar]
- [25].Primeau AJ, Rendon A, Hedley D, Lilge L, Tannock IF. The distribution of the anticancer drug Doxorubicin in relation to blood vessels in solid tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11:8782–8788. doi: 10.1158/1078-0432.CCR-05-1664. [DOI] [PubMed] [Google Scholar]
- [26].Saggar JK, Yu M, Tan Q, Tannock IF. The tumor microenvironment and strategies to improve drug distribution. Frontiers in oncology. 2013;3:154. doi: 10.3389/fonc.2013.00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Folkman J. Tumor angiogenesis. Advances in Cancer Research. 1985;43:175–203. doi: 10.1016/s0065-230x(08)60946-x. [DOI] [PubMed] [Google Scholar]
- [28].Folkman J, Bach M, Rowe JW, Davidoff F, Lambert P, Hirsch C, Goldberg A, Hiatt HH, Glass J, Henshaw E. Tumor angiogenesis- therapeutic implications. New England Journal of Medicine. 1971;285:1182. doi: 10.1056/NEJM197111182852108. &. [DOI] [PubMed] [Google Scholar]
- [29].Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, Roberge S, Jain RK, McDonald DM. Openings between defective endothelial cells explain tumor vessel leakiness. American Journal of Pathology. 2000;156:1363–1380. doi: 10.1016/S0002-9440(10)65006-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Hobbs SK, Monsky WL, Yuan F, Roberts WG, Griffith L, Torchilin VP, Jain RK. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:4607–4612. doi: 10.1073/pnas.95.8.4607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Dvorak HF, Nagy JA, Dvorak JT, Dvorak AM. Identification and characterization of the blood-vessels of solid tumors that are leaky to circulating macromolecules. American Journal of Pathology. 1988;133:95–109. [PMC free article] [PubMed] [Google Scholar]
- [32].Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular-permeability factor/vascular endothelial growth-factor, microvascular hyperpermeability, and angiogenesis. American Journal of Pathology. 1995;146:1029–1039. [PMC free article] [PubMed] [Google Scholar]
- [33].Roberts WG, Palade GE. Neovasculature induced by vascular endothelial growth factor is fenestrated. Cancer Research. 1997;57:765–772. [PubMed] [Google Scholar]
- [34].Jain RK. Determinants of tumor blood-flow - a review. Cancer Research. 1988;48:2641–2658. [PubMed] [Google Scholar]
- [35].Nagy JA, Benjamin L, Zeng H, Dvorak AM, Dvorak HF. Vascular permeability, vascular hyperpermeability and angiogenesis. Angiogenesis. 2008;11:109–119. doi: 10.1007/s10456-008-9099-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Nagy JA, Chang S-H, Shih S-C, Dvorak AM, Dvorak HF. Heterogeneity of the Tumor Vasculature. Seminars in Thrombosis and Hemostasis. 2010;36:321–331. doi: 10.1055/s-0030-1253454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Jain RK. Transport of molecules across tumor vasculature. Cancer and Metastasis Reviews. 1987;6:559–593. doi: 10.1007/BF00047468. [DOI] [PubMed] [Google Scholar]
- [38].Harris AL. Hypoxia - A key regulatory factor in tumour growth. Nat. Rev. Cancer. 2002;2:38–47. doi: 10.1038/nrc704. [DOI] [PubMed] [Google Scholar]
- [39].Hockel M, Vaupel P. Tumor hypoxia: Definitions and current clinical, biologic, and molecular aspects. Journal of the National Cancer Institute. 2001;93:266–276. doi: 10.1093/jnci/93.4.266. [DOI] [PubMed] [Google Scholar]
- [40].Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO(2) gradients in solid tumors in vivo: High-resolution measurements reveal a lack of correlation. Nature medicine. 1997;3:177–182. doi: 10.1038/nm0297-177. [DOI] [PubMed] [Google Scholar]
- [41].Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer. 2004;4:891–899. doi: 10.1038/nrc1478. [DOI] [PubMed] [Google Scholar]
- [42].Semenza GL. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer. 2003;3:721–732. doi: 10.1038/nrc1187. [DOI] [PubMed] [Google Scholar]
- [43].Fukumura D, Xu L, Chen Y, Gohongi T, Seed B, Jain RK. Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Research. 2001;61:6020–6024. [PubMed] [Google Scholar]
- [44].Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza GL. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and Cellular Biology. 1996;16:4604–4613. doi: 10.1128/mcb.16.9.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Majmundar AJ, Wong WJ, Simon MC. Hypoxia-Inducible Factors and the Response to Hypoxic Stress. Molecular Cell. 2010;40:294–309. doi: 10.1016/j.molcel.2010.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Huang LE, Gu J, Schau M, Bunn HF. Regulation of hypoxia-inducible factor 1 alpha is mediated by an O-2-dependent degradation domain via the ubiquitin-proteasome pathway. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:7987–7992. doi: 10.1073/pnas.95.14.7987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–275. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
- [48].Veikkola T, Karkkainen M, Claesson-Welsh L, Alitalo K. Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Research. 2000;60:203–212. [PubMed] [Google Scholar]
- [49].Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nature medicine. 2003;9:669–676. doi: 10.1038/nm0603-669. [DOI] [PubMed] [Google Scholar]
- [50].Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nature reviews. Cancer. 2003;3:401–410. doi: 10.1038/nrc1093. [DOI] [PubMed] [Google Scholar]
- [51].Saggar JK, Fung AS, Patel KJ, Tannock IF. Use of Molecular Biomarkers to Quantify the Spatial Distribution of Effects of Anticancer Drugs in Solid Tumors. Molecular Cancer Therapeutics. 2013;12:542–552. doi: 10.1158/1535-7163.MCT-12-0967. [DOI] [PubMed] [Google Scholar]
- [52].Jain RK. Transport of molecules in the tumor interstitium- a review. Cancer Research. 1987;47:3039–3051. [PubMed] [Google Scholar]
- [53].Baxter LT, Jain RK. Transport of fluid and macromolecules in tumors 1. Role of interstitial pressure and convection. Microvascular research. 1989;37:77–104. doi: 10.1016/0026-2862(89)90074-5. [DOI] [PubMed] [Google Scholar]
- [54].Primeau AJ, Rendon A, Hedley D, Lilge L, Tannock IF. The distribution of the anticancer drug Doxorubicin in relation to blood vessels in solid tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11:8782–8788. doi: 10.1158/1078-0432.CCR-05-1664. [DOI] [PubMed] [Google Scholar]
- [55].Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nature reviews. Cancer. 2006;6:583–592. doi: 10.1038/nrc1893. [DOI] [PubMed] [Google Scholar]
- [56].Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nature medicine. 2001;7:987–989. doi: 10.1038/nm0901-987. [DOI] [PubMed] [Google Scholar]
- [57].Jain RK. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science. 2005;307:58–62. doi: 10.1126/science.1104819. [DOI] [PubMed] [Google Scholar]
- [58].Fukumura D, Jain RK. Tumor microvasculature and microenvironment: Targets for anti-angiogenesis and normalization. Microvascular Research. 2007;74:72–84. doi: 10.1016/j.mvr.2007.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nature Reviews Drug Discovery. 2011;10:417–427. doi: 10.1038/nrd3455. [DOI] [PubMed] [Google Scholar]
- [60].Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK. Normalization of the vasculature for treatment of cancer and other diseases. Physiological Reviews. 2011;91:1071–1121. doi: 10.1152/physrev.00038.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat. Rev. Cancer. 2008;8:579–591. doi: 10.1038/nrc2403. [DOI] [PubMed] [Google Scholar]
- [62].Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. New England Journal of Medicine. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
- [63].Nathanson SD, Nelson L. Interstitial Fluid Pressure in Breast-Cancer, Benign Breast Conditions, and Breast Parenchyma. Ann Surg Oncol. 1994;1:333–338. doi: 10.1007/BF03187139. [DOI] [PubMed] [Google Scholar]
- [64].Less JR, Posner MC, Boucher Y, Borochovitz D, Wolmark N, Jain RK. Interstitial Hypertension in Human Tumors .4. Interstitial Hypertension in Human Breast and Colorectal Tumors. Cancer Research. 1992;52:6371–6374. [PubMed] [Google Scholar]
- [65].Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure - An obstacle in cancer therapy. Nat. Rev. Cancer. 2004;4:806–813. doi: 10.1038/nrc1456. [DOI] [PubMed] [Google Scholar]
- [66].Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nature Reviews Molecular Cell Biology. 2006;7:211–224. doi: 10.1038/nrm1858. [DOI] [PubMed] [Google Scholar]
- [67].Truskey FYGA, Katz DF. Pearson Prentice Hall. Transport phenomena in biological systems. 2004 [Google Scholar]
- [68].Thurber GM, Schmidt MM, Wittrup KD. Factors determining antibody distribution in tumors. Trends in Pharmacological Sciences. 2008;29:57–61. doi: 10.1016/j.tips.2007.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Gauvin R, Guillemette M, Dokmeci M, Khademhosseini A. Application of microtechnologies for the vascularization of engineered tissues. Vascular cell. 2011;3:24–24. doi: 10.1186/2045-824X-3-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Young EWK. Cells, tissues, and organs on chips: challenges and opportunities for the cancer tumor microenvironment. Integrative Biology. 2013;5:1096–1109. doi: 10.1039/c3ib40076j. [DOI] [PubMed] [Google Scholar]
- [71].Stroock AD, Fischbach C. Microfluidic Culture Models of Tumor Angiogenesis. Tissue Engineering Part A. 2010;16:2143–2146. doi: 10.1089/ten.tea.2009.0689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Song JW, Munn LL. Fluid forces control endothelial sprouting. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:15342–15347. doi: 10.1073/pnas.1105316108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Shin Y, Han S, Jeon JS, Yamamoto K, Zervantonakis IK, Sudo R, Kamm RD, Chung S. Microfluidic assay for simultaneous culture of multiple cell types on surfaces or within hydrogels. Nature protocols. 2012;7:1247–1259. doi: 10.1038/nprot.2012.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Kim S, Lee H, Chung M, Jeon NL. Engineering of functional, perfusable 3D microvascular networks on a chip. Lab on a chip. 2013;13:1489–1500. doi: 10.1039/c3lc41320a. [DOI] [PubMed] [Google Scholar]
- [75].Chrobak KM, Potter DR, Tien J. Formation of perfused, functional microvascular tubes in vitro. Microvascular Research. 2006;71:185–196. doi: 10.1016/j.mvr.2006.02.005. [DOI] [PubMed] [Google Scholar]
- [76].Nguyen D-HT, Stapleton SC, Yang MT, Cha SS, Choi CK, Galie PA, Chen CS. Biomimetic model to reconstitute angiogenic sprouting morphogenesis in vitro. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:6712–6717. doi: 10.1073/pnas.1221526110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Golden AP, Tien J. Fabrication of microfluidic hydrogels using molded gelatin as a sacrificial element. Lab on a Chip. 2007;7:720–725. doi: 10.1039/b618409j. [DOI] [PubMed] [Google Scholar]
- [78].Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT, Cohen DM, Toro E, Chen A.a., Galie P.a., Yu X, Chaturvedi R, Bhatia SN, Chen CS. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nature materials. 2012;11:768–774. doi: 10.1038/nmat3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Whitesides GM, Ostuni E, Takayama S, Jiang XY, Ingber DE. Soft lithography in biology and biochemistry. Annual review of biomedical engineering. 2001;3:335–373. doi: 10.1146/annurev.bioeng.3.1.335. [DOI] [PubMed] [Google Scholar]
- [80].Borenstein JT, Terai H, King KR, Weinberg EJ, Kaazempur-Mofrad MR, Vacanti JP. Microfabrication technology for vascularized tissue engineering. Biomedical microdevices. 2002;4:167–175. [Google Scholar]
- [81].Choi NW, Cabodi M, Held B, Gleghorn JP, Bonassar LJ, Stroock AD. Microfluidic scaffolds for tissue engineering. Nature Materials. 2007;6:908–915. doi: 10.1038/nmat2022. [DOI] [PubMed] [Google Scholar]
- [82].Zheng Y, Chen J, Craven M, Choi NW, Totorica S, Diaz-Santana A, Kermani P, Hempstead B, Fischbach-Teschl C, Lopez JA, Stroock AD. In vitro microvessels for the study of angiogenesis and thrombosis. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:9342–9347. doi: 10.1073/pnas.1201240109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Verbridge SS, Chakrabarti A, DelNero P, Kwee B, Varner J, Stroock AD, Fischbach C. Physicochemical regulation of endothelial sprouting in a 3D microfluidic angiogenesis model. Journal of Biomedical Materials Research Part A. 2013;10:2948–2956. doi: 10.1002/jbm.a.34587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].McGuigan AP, Sefton MV. Vascularized organoid engineered by modular assembly enables blood perfusion. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:11461–11466. doi: 10.1073/pnas.0602740103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Du Y, Ghodousi M, Qi H, Haas N, Xiao W, Khademhosseini A. Sequential assembly of cell-laden hydrogel constructs to engineer vascular-like microchannels. Biotechnology and bioengineering. 2011;108:1693–1703. doi: 10.1002/bit.23102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Wong KHK, Chan JM, Kamm RD, Tien J. Microfluidic Models of Vascular Functions. Annual Review of Biomedical Engineering. 2012;Vol 14:205–230. doi: 10.1146/annurev-bioeng-071811-150052. 14. [DOI] [PubMed] [Google Scholar]
- [87].Huang CP, Lu J, Seon H, Lee AP, Flanagan LA, Kim HY, Putnam AJ, Jeon NL. Engineering microscale cellular niches for three-dimensional multicellular co-cultures. Lab on a chip. 2009;9:1740–1748. doi: 10.1039/b818401a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Sung KE, Yang N, Pehlke C, Keely PJ, Eliceiri KW, Friedl A, Beebe DJ. Transition to invasion in breast cancer: a microfluidic in vitro model enables examination of spatial and temporal effects. Integrative Biology. 2011;3:439–450. doi: 10.1039/c0ib00063a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Moya M, Tran D, George S. An integrated in vitro model of perfused tumor and cardiac tissue. Stem Cell Research and Therapy. 2013;4 doi: 10.1186/scrt376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Sung KE, Su X, Berthier E, Pehlke C, Friedl A, Beebe D. Understanding the impact of 2D and 3D fibroblast cultures on in vitro breast cancer models. PloS one. 2013;8 doi: 10.1371/journal.pone.0076373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. International journal of cancer. Journal international du cancer. 2008;123:2229–2238. doi: 10.1002/ijc.23925. [DOI] [PubMed] [Google Scholar]
- [92].Egeblad M, Rasch MG, Weaver VM. Dynamic interplay between the collagen scaffold and tumor evolution. Current opinion in cell biology. 2010;22:697–706. doi: 10.1016/j.ceb.2010.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech. 2011;4:165–178. doi: 10.1242/dmm.004077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nature reviews. Cancer. 2009;9:108–122. doi: 10.1038/nrc2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Baneyx G, Baugh L, Vogel V. Fibronectin extension and unfolding within cell matrix fibrils controlled by cytoskeletal tension. Proc Natl Acad Sci U S A. 2002;99:5139–5143. doi: 10.1073/pnas.072650799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Chandler EM, Saunders MP, Yoon CJ, Gourdon D, Fischbach C. Adipose progenitor cells increase fibronectin matrix strain and unfolding in breast tumors. Physical biology. 2011;8:015008. doi: 10.1088/1478-3975/8/1/015008. [DOI] [PubMed] [Google Scholar]
- [97].Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3:349–363. doi: 10.1038/nrm809. [DOI] [PubMed] [Google Scholar]
- [98].Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139:891–906. doi: 10.1016/j.cell.2009.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clinical & experimental metastasis. 2009;26:35–49. doi: 10.1007/s10585-008-9209-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141:52–67. doi: 10.1016/j.cell.2010.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Davis K.J.B. George E., Davis Michael J., Meininger Gerald A. Regulation of Tissue Injury Responses by the Exposure of Matricryptic Sites within Extracellular Matrix Molecules. American Journal of Pathology. 2000;156:1489–1498. doi: 10.1016/S0002-9440(10)65020-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Wolf K, Wu YI, Liu Y, Geiger J, Tam E, Overall C, Stack MS, Friedl P. Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nature cell biology. 2007;9:893–904. doi: 10.1038/ncb1616. [DOI] [PubMed] [Google Scholar]
- [103].Johnson C, Galis ZS. Matrix metalloproteinase-2 and -9 differentially regulate smooth muscle cell migration and cell-mediated collagen organization. Arterioscler Thromb Vasc Biol. 2004;24:54–60. doi: 10.1161/01.ATV.0000100402.69997.C3. [DOI] [PubMed] [Google Scholar]
- [104].Defawe OD, Kenagy RD, Choi C, Wan SYC, Deroanne C, Nusgens B, Sakalihasan N, Colige A, Clowes AW. MMP-9 regulates both positively and negatively collagen gel contraction - A nonproteolytic function of MMP-9. Cardiovasc Res. 2005;66:402–409. doi: 10.1016/j.cardiores.2004.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer. 2002;2:161–174. doi: 10.1038/nrc745. [DOI] [PubMed] [Google Scholar]
- [106].Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nature Reviews Molecular Cell Biology. 2007;8:221–233. doi: 10.1038/nrm2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK-ERK linkage. Oncogene. 2009;28:4326–4343. doi: 10.1038/onc.2009.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Vogel V, Sheetz M. Local force and geometry sensing regulate cell functions. Nat Rev Mol Cell Biol. 2006;7:265–275. doi: 10.1038/nrm1890. [DOI] [PubMed] [Google Scholar]
- [109].Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5:816–826. doi: 10.1038/nrm1490. [DOI] [PubMed] [Google Scholar]
- [110].Fischbach C, Kong HJ, Hsiong SX, Evangelista MB, Yuen W, Mooney DJ. Cancer cell angiogenic capability is regulated by 3D culture and integrin engagement. Proc Natl Acad Sci U S A. 2009;106:399–404. doi: 10.1073/pnas.0808932106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Geiger B, Spatz JP, Bershadsky AD. Environmental sensing through focal adhesions. Nat Rev Mol Cell Biol. 2009;10:21–33. doi: 10.1038/nrm2593. [DOI] [PubMed] [Google Scholar]
- [112].Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–689. doi: 10.1016/j.cell.2006.06.044. [DOI] [PubMed] [Google Scholar]
- [113].Schrader J, Gordon-Walker TT, Aucott RL, van Deemter M, Quaas A, Walsh S, Benten D, Forbes SJ, Wells RG, Iredale JP. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology. 2011;53:1192–1205. doi: 10.1002/hep.24108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Chandler EM, Seo BR, Califano JP, Eguiluz R.C. Andresen, Lee JS, Yoon CJ, Tims DT, Wang JX, Cheng L, Mohanan S, Buckley MR, Cohen I, Nikitin AY, Williams RM, Gourdon D, Reinhart-King CA, Fischbach C. Implanted adipose progenitor cells as physicochemical regulators of breast cancer. Proc Natl Acad Sci U S A. 2012;109:9786–9791. doi: 10.1073/pnas.1121160109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Castello-Cros R, Khan DR, Simons J, Valianou M, Cukierman E. Staged stromal extracellular 3D matrices differentially regulate breast cancer cell responses through PI3K and beta1-integrins. BMC cancer. 2009;9:94. doi: 10.1186/1471-2407-9-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].DuFort CC, Paszek MJ, Weaver VM. Balancing forces: architectural control of mechanotransduction. Nat Rev Mol Cell Biol. 2011;12:308–319. doi: 10.1038/nrm3112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Dienstmann R, De Dosso S, Felip E, Tabernero J. Drug development to overcome resistance to EGFR inhibitors in lung and colorectal cancer. Molecular oncology. 2012;6:15–26. doi: 10.1016/j.molonc.2011.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Paraiso KH, Xiang Y, Rebecca VW, Abel EV, Chen YA, Munko AC, Wood E, Fedorenko IV, Sondak VK, Anderson AR, Ribas A, Palma MD, Nathanson KL, Koomen JM, Messina JL, Smalley KS. PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression. Cancer Res. 2011;71:2750–2760. doi: 10.1158/0008-5472.CAN-10-2954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Sottile J. Regulation of angiogenesis by extracellular matrix. Biochimica et biophysica acta. 2004;1654:13–22. doi: 10.1016/j.bbcan.2003.07.002. [DOI] [PubMed] [Google Scholar]
- [120].Huebsch N, Arany PR, Mao AS, Shvartsman D, Ali OA, Bencherif SA, Rivera-Feliciano J, Mooney DJ. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat Mater. 2010;9:518–526. doi: 10.1038/nmat2732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Hammer DA, Weaver VM. Tensional homeostasis and the malignant phenotype. Cancer cell. 2005;8:241–254. doi: 10.1016/j.ccr.2005.08.010. [DOI] [PubMed] [Google Scholar]
- [122].Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, Andreeff M, Marini F. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PloS one. 2009;4:e4992. doi: 10.1371/journal.pone.0004992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Mammoto A, Mammoto T, Kanapathipillai M, Yung C. Wing, Jiang E, Jiang A, Lofgren K, Gee EP, Ingber DE. Control of lung vascular permeability and endotoxin-induced pulmonary oedema by changes in extracellular matrix mechanics. Nature communications. 2013;4:1759. doi: 10.1038/ncomms2774. [DOI] [PubMed] [Google Scholar]
- [124].Huynh J, Nishimura N, Rana K, Peloquin JM, Califano JP, Montague CR, King MR, Schaffer CB, Reinhart-King CA. Age-related intimal stiffening enhances endothelial permeability and leukocyte transmigration. Science translational medicine. 2011;3:112ra122. doi: 10.1126/scitranslmed.3002761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Patel M.B. Naimish R., Chen Cheng, Hardin Charles C., Kho Alvin T., Mih Justin, Deng Linhong, Butler James, Tschumperlin Daniel, Fredberg Jeffrey J., Krishnan Ramaswamy, Koziel Henry. Cell Elasticity Determines Macrophage Function. PloS one. 2012;7 doi: 10.1371/journal.pone.0041024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen HY, Omeroglu G, Meterissian S, Omeroglu A, Hallett M, Park M. Stromal gene expression predicts clinical outcome in breast cancer. Nature medicine. 2008;14:518–527. doi: 10.1038/nm1764. [DOI] [PubMed] [Google Scholar]
- [127].Kellermann MG, Sobral LM, Silva S.D. da, Zecchin KG, Graner E, Lopes MA, Nishimoto I, Kowalski LP, Coletta RD. Myofibroblasts in the stroma of oral squamous cell carcinoma are associated with poor prognosis. Histopathology. 2007;51:849–853. doi: 10.1111/j.1365-2559.2007.02873.x. [DOI] [PubMed] [Google Scholar]
- [128].Caporale A, Bonapasta SA, Scarpini M, Ciardi A, Vestri A, Ruperto M, Giuliani A. Quantitative Investigation of Desmoplasia as a Prognostic Indicator in Colorectal Cancer. J Invest Surg. 2010;23:105–109. doi: 10.3109/08941930903469417. [DOI] [PubMed] [Google Scholar]
- [129].Hasebe T, Tsuda H, Hirohashi S, Shimosato Y, Iwai M, Imoto S, Mukai K. Fibrotic focus in invasive ductal carcinoma: An indicator of high tumor aggressiveness. Jpn J Cancer Res. 1996;87:385–394. doi: 10.1111/j.1349-7006.1996.tb00234.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Van den Eynden GG, Colpaert CG, Couvelard A, Pezzella F, Dirix LY, Vermeulen PB, Van Marck EA, Hasebe T. A fibrotic focus is a prognostic factor and a surrogate marker for hypoxia and (lymph)angiogenesis in breast cancer: review of the literature and proposal on the criteria of evaluation. Histopathology. 2007;51:440–451. doi: 10.1111/j.1365-2559.2007.02761.x. [DOI] [PubMed] [Google Scholar]
- [131].Halvorsen TB, Seim E. Association between Invasiveness, Inflammatory Reaction, Desmoplasia and Survival in Colorectal-Cancer. J Clin Pathol. 1989;42:162–166. doi: 10.1136/jcp.42.2.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Karagiannis GS, Petraki C, Prassas I, Saraon P, Musrap N, Dimitromanolakis A, Diamandis EP. Proteomic Signatures of the Desmoplastic Invasion Front Reveal Collagen Type XII as a Marker of Myofibroblastic Differentiation During Colorectal Cancer Metastasis. Oncotarget. 2012;3:267–285. doi: 10.18632/oncotarget.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Okada KI, Hirabayashi K, Imaizumi T, Matsuyama M, Yazawa N, Dowaki S, Tobita K, Ohtani Y, Tanaka M, Inokuchi S, Makuuchi H. Stromal thrombospondin-1 expression is a prognostic indicator and a new marker of invasiveness in intraductal papillary-mucinous neoplasm of the pancreas. Biomed Res-Tokyo. 2010;31:13–19. doi: 10.2220/biomedres.31.13. [DOI] [PubMed] [Google Scholar]
- [134].Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nature reviews. Molecular cell biology. 2006;7:211–224. doi: 10.1038/nrm1858. [DOI] [PubMed] [Google Scholar]
- [135].Mitsi M, Hong Z, Costello CE, Nugent MA. Heparin-mediated conformational changes in fibronectin expose Vascular Endothelial Growth Factor binding sites. Biochemistry. 2006;45:10319–10328. doi: 10.1021/bi060974p. [DOI] [PubMed] [Google Scholar]
- [136].Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Current opinion in cell biology. 2004;16:558–564. doi: 10.1016/j.ceb.2004.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Bouzin C, Feron O. Targeting tumor stroma and exploiting mature tumor vasculature to improve anti-cancer drug delivery. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy. 2007;10:109–120. doi: 10.1016/j.drup.2007.03.001. [DOI] [PubMed] [Google Scholar]
- [138].Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure - an obstacle in cancer therapy. Nature reviews. Cancer. 2004;4:806–813. doi: 10.1038/nrc1456. [DOI] [PubMed] [Google Scholar]
- [139].Pluen A, Boucher Y, Ramanujan S, McKee TD, Gohongi T, di Tomaso E, Brown EB, Izumi Y, Campbell RB, Berk DA, Jain RK. Role of tumor-host interactions in interstitial diffusion of macromolecules: cranial vs. subcutaneous tumors. Proc Natl Acad Sci U S A. 2001;98:4628–4633. doi: 10.1073/pnas.081626898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Chauhan VP, Lanning RM, Diop-Frimpong B, Mok W, Brown EB, Padera TP, Boucher Y, Jain RK. Multiscale measurements distinguish cellular and interstitial hindrances to diffusion in vivo. Biophysical journal. 2009;97:330–336. doi: 10.1016/j.bpj.2009.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Ramanujan S, Pluen A, McKee TD, Brown EB, Boucher Y, Jain RK. Diffusion and convection in collagen gels: implications for transport in the tumor interstitium. Biophysical journal. 2002;83:1650–1660. doi: 10.1016/S0006-3495(02)73933-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Netti PA, Berk DA, Swartz MA, Grodzinsky AJ, Jain RK. Role of Extracellular Matrix Assembly in Interstitial Transport in Solid Tumors. Cancer Res. 2000;60:2497–2503. [PubMed] [Google Scholar]
- [143].Choi HS, Liu WH, Liu FB, Nasr K, Misra P, Bawendi MG, Frangioni JV. Design considerations for tumour-targeted nanoparticles. Nature Nanotechnology. 2010;5:42–47. doi: 10.1038/nnano.2009.314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Stylianopoulos T, Poh MZ, Insin N, Bawendi MG, Fukumura D, Munn LL, Jain RK. Diffusion of particles in the extracellular matrix: the effect of repulsive electrostatic interactions. Biophysical journal. 2010a;99:1342–1349. doi: 10.1016/j.bpj.2010.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Yuan F, Krol A, Tong S. Available Space and Extracellular Transport of Macromolecules: Effects of Pore Size and Connectedness. Annals of Biomedical Engineering. 2001;29:1150–1158. doi: 10.1114/1.1424915. [DOI] [PubMed] [Google Scholar]
- [146].Choi J, Credit K, Henderson K, Deverkadra R, He Z, Wiig H, Vanpelt H, Flessner MF. Intraperitoneal immunotherapy for metastatic ovarian carcinoma: Resistance of intratumoral collagen to antibody penetration. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12:1906–1912. doi: 10.1158/1078-0432.CCR-05-2141. [DOI] [PubMed] [Google Scholar]
- [147].Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC medicine. 2006;4:38. doi: 10.1186/1741-7015-4-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, White JG, Keely PJ. Collagen density promotes mammary tumor initiation and progression. BMC medicine. 2008;6 doi: 10.1186/1741-7015-6-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Stylianopoulos T, Diop-Frimpong B, Munn LL, Jain RK. Diffusion anisotropy in collagen gels and tumors: the effect of fiber network orientation. Biophysical journal. 2010b;99:3119–3128. doi: 10.1016/j.bpj.2010.08.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Lieleg O, Baumgartel RM, Bausch AR. Selective filtering of particles by the extracellular matrix: an electrostatic bandpass. Biophysical journal. 2009;97:1569–1577. doi: 10.1016/j.bpj.2009.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Wan AM, Schur RM, Ober CK, Fischbach C, Gourdon D, Malliaras GG. Electrical control of protein conformation. Advanced materials. 2012;24:2501–2505. doi: 10.1002/adma.201200436. [DOI] [PubMed] [Google Scholar]
- [152].Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, DeNicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Rueckert F, Gruetzmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, Hruban RH, Whitebread N, McGovern K, Adams J, Iacobuzio-Donahue C, Griffiths J, Tuveson DA. Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer. Science. 2009;324:1457–1461. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol. 2005;15:378–386. doi: 10.1016/j.semcancer.2005.05.004. [DOI] [PubMed] [Google Scholar]
- [154].Clarke KC, Douglas AM, Brown AC, Barker TH, Lyon LA. Colloid-matrix assemblies in regenerative medicine. Current Opinion in Colloid & Interface Science. 2013;18:393–405. [Google Scholar]
- [155].Sodek KL, Brown TJ, Ringuette MJ. Collagen I but not Matrigel matrices provide an MMP-dependent barrier to ovarian cancer cell penetration. BMC cancer. 2008;8:223. doi: 10.1186/1471-2407-8-223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Girton TS, Oegema TR, Tranquillo RT. Exploiting glycation to stiffen and strengthen tissue equivalents for tissue engineering. Journal of Biomedical Materials Research. 1999;46:87–92. doi: 10.1002/(sici)1097-4636(199907)46:1<87::aid-jbm10>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
- [157].Goldin A, Beckman JA, Schmidt AM, Creager MA. Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation. 2006;114:597–605. doi: 10.1161/CIRCULATIONAHA.106.621854. [DOI] [PubMed] [Google Scholar]
- [158].Fraley SI, Feng Y, Krishnamurthy R, Kim DH, Celedon A, Longmore GD, Wirtz D. A distinctive role for focal adhesion proteins in three-dimensional cell motility. Nature cell biology. 2010;12:598–604. doi: 10.1038/ncb2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Trappmann B, Gautrot JE, Connelly JT, Strange DG, Li Y, Oyen ML, Stuart M.A. Cohen, Boehm H, Li B, Vogel V, Spatz JP, Watt FM, Huck WT. Extracellular-matrix tethering regulates stem-cell fate. Nat Mater. 2012;11:642–649. doi: 10.1038/nmat3339. [DOI] [PubMed] [Google Scholar]
- [160].Gill BJ, Gibbons DL, Roudsari LC, Saik JE, Rizvi ZH, Roybal JD, Kurie JM, West JL. A synthetic matrix with independently tunable biochemistry and mechanical properties to study epithelial morphogenesis and EMT in a lung adenocarcinoma model. Cancer Res. 2012;72:6013–6023. doi: 10.1158/0008-5472.CAN-12-0895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Kloxin AM, Kasko AM, Salinas CN, Anseth KS. Photodegradable hydrogels for dynamic tuning of physical and chemical properties. Science. 2009;324:59–63. doi: 10.1126/science.1169494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].DeForest CA, Anseth KS. Photoreversible patterning of biomolecules within click-based hydrogels. Angewandte Chemie. 2012;51:1816–1819. doi: 10.1002/anie.201106463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Khetan S, Guvendiren M, Legant WR, Cohen DM, Chen CS, Burdick JA. Degradation-mediated cellular traction directs stem cell fate in covalently crosslinked three-dimensional hydrogels. Nat Mater. 2013;12:458–465. doi: 10.1038/nmat3586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature biotechnology. 2005;23:47–55. doi: 10.1038/nbt1055. [DOI] [PubMed] [Google Scholar]
- [165].Achilli M, Mantovani D. Tailoring Mechanical Properties of Collagen-Based Scaffolds for Vascular Tissue Engineering: The Effects of pH, Temperature and Ionic Strength on Gelation. Polymers. 2010;2:664–680. [Google Scholar]
- [166].Zeugolis DI, Paul RG, Attenburrow G. Factors influencing the properties of reconstituted collagen fibers prior to self-assembly: animal species and collagen extraction method. Journal of biomedical materials research. Part A. 2008;86:892–904. doi: 10.1002/jbm.a.31694. [DOI] [PubMed] [Google Scholar]
- [167].Friess W. Collagen-biomaterial for drug delivery. European Journal of Pharmaceutics and Biopharmaceutics. 1998;45:113–136. doi: 10.1016/s0939-6411(98)00017-4. [DOI] [PubMed] [Google Scholar]
- [168].Brownfield DG, Venugopalan G, Lo A, Mori H, Tanner K, Fletcher DA, Bissell MJ. Patterned collagen fibers orient branching mammary epithelium through distinct signaling modules. Current biology : CB. 2013;23:703–709. doi: 10.1016/j.cub.2013.03.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Moraes C, Sun Y, Simmons CA. (Micro)managing the mechanical microenvironment. Integrative biology : quantitative biosciences from nano to macro. 2011;3:959–971. doi: 10.1039/c1ib00056j. [DOI] [PubMed] [Google Scholar]
- [170].W.G. Matthews JA, Simpson DG, Bowlin GL. Electrospinning of collagen nanofibers. Biomacromolecules. 2002;3:232–238. doi: 10.1021/bm015533u. [DOI] [PubMed] [Google Scholar]
- [171].Kim TG, Park TG. Biomimicking Extracellular Matrix: Cell Adhesive RGD Peptide Modified Electrospun Poly(D,L-lactic-co-glycolic acid) Nanofiber Mesh. Tissue Engineering. 2006;12:221–233. doi: 10.1089/ten.2006.12.221. [DOI] [PubMed] [Google Scholar]
- [172].Jang JH, Castano O, Kim HW. Electrospun materials as potential platforms for bone tissue engineering. Advanced drug delivery reviews. 2009;61:1065–1083. doi: 10.1016/j.addr.2009.07.008. [DOI] [PubMed] [Google Scholar]
- [173].Hasan A, Memic A, Annabi N, Hossain M, Paul A, Dokmeci MR, Dehghani F, Khademhosseini A. Electrospun scaffolds for tissue engineering of vascular grafts. Acta biomaterialia. 2013 doi: 10.1016/j.actbio.2013.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Smith ML, Gourdon D, Little WC, Kubow KE, Eguiluz RA, Luna-Morris S, Vogel V. Force-induced unfolding of fibronectin in the extracellular matrix of living cells. PLoS biology. 2007;5:e268. doi: 10.1371/journal.pbio.0050268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Brown AC, Rowe JA, Barker TH. Guiding epithelial cell phenotypes with engineered integrin-specific recombinant fibronectin fragments. Tissue engineering. Part A. 2011;17:139–150. doi: 10.1089/ten.tea.2010.0199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Wan AM, Brooks DJ, Gumus A, Fischbach C, Malliaras GG. Electrical control of cell density gradients on a conducting polymer surface. Chemical communications. 2009:5278–5280. doi: 10.1039/b911130a. [DOI] [PubMed] [Google Scholar]
- [177].Wan AM, Chandler EM, Madhavan M, Infanger DW, Ober CK, Gourdon D, Malliaras GG, Fischbach C. Fibronectin conformation regulates the proangiogenic capability of tumor-associated adipogenic stromal cells. Biochimica et biophysica acta. 2013;1830:4314–4320. doi: 10.1016/j.bbagen.2013.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Amatangelo MD, Bassi DE, Klein-Szanto AJP, Cukierman E. Stroma-derived three-dimensional matrices are necessary and sufficient to promote desmoplastic differentiation of normal fibroblasts. American Journal of Pathology. 2005;167:475–488. doi: 10.1016/S0002-9440(10)62991-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Serebriiskii I, Castello-Cros R, Lamb A, Golemis EA, Cukierman E. Fibroblast-derived 3D matrix differentially regulates the growth and drug-responsiveness of human cancer cells. Matrix Biol. 2008;27:573–585. doi: 10.1016/j.matbio.2008.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Gobaa S, Hoehnel S, Roccio M, Negro A, Kobel S, Lutolf MP. Artificial niche microarrays for probing single stem cell fate in high throughput. Nature methods. 2011;8:949–955. doi: 10.1038/nmeth.1732. [DOI] [PubMed] [Google Scholar]
- [181].Paguirigan AL, Beebe DJ. From the cellular perspective: exploring differences in the cellular baseline in macroscale and microfluidic cultures. Integrative Biology. 2009;1:182–195. doi: 10.1039/b814565b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Regehr KJ, Domenech M, Koepsel JT, Carver KC, Ellison-Zelski SJ, Murphy WL, Schuler LA, Alarid ET, Beebe DJ. Biological implications of polydimethylsiloxane-based microfluidic cell culture. Lab on a Chip. 2009;9:2132–2139. doi: 10.1039/b903043c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab on a chip. 2006;6:1484–1486. doi: 10.1039/b612140c. [DOI] [PubMed] [Google Scholar]
- [184].Sung JH, Shuler ML. A micro cell culture analog (mu CCA) with 3-D hydrogel culture of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer drugs. Lab on a chip. 2009;9:1385–1394. doi: 10.1039/b901377f. [DOI] [PubMed] [Google Scholar]
- [185].Sung JH, Kam C, Shuler ML. A microfluidic device for a pharmacokinetic-pharmacodynamic (PK-PD) model on a chip. Lab on a chip. 2010;10:446–455. doi: 10.1039/b917763a. [DOI] [PubMed] [Google Scholar]
- [186].Kaplan RN, Rafii S, Lyden D. Preparing the “Soil”: The premetastatic niche. Cancer Research. 2006;66:11089–11093. doi: 10.1158/0008-5472.CAN-06-2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Esch MB, King TL, Shuler ML. The Role of Body-on-a-Chip Devices in Drug and Toxicity Studies. Annual Review of Biomedical Engineering. 2011;Vol 13:55–72. doi: 10.1146/annurev-bioeng-071910-124629. 13. [DOI] [PubMed] [Google Scholar]
- [188].DelNero P, Song YH, Fischbach C. Biomedical Microdevices. 2013. Microengineered tumor models: insights and opportunities from a physical sciences-oncology perspective; pp. 583–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Gerlinger M, Swanton C. How Darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine. British journal of cancer. 2010;103:1139–1143. doi: 10.1038/sj.bjc.6605912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. New England Journal of Medicine. 2012;366:883–892. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Carey SP, Starchenko A, McGregor AL, Reinhart-King CA. Leading malignant cells initiate collective epithelial cell invasion in a three-dimensional heterotypic tumor spheroid model. Clinical & experimental metastasis. 2013;30:615–630. doi: 10.1007/s10585-013-9565-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Yang S, Graham J, Kahn JW, Schwartz EA, Gerritsen ME. Functional roles for PECAM-1 (CD31) and VE-cadherin (CD144) in tube assembly and lumen formation in three-dimensional collagen gels. American Journal of Pathology. 1999;155:887–895. doi: 10.1016/S0002-9440(10)65188-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Sieminski AL, Hebbel RP, Gooch KJ. Improved microvascular network in vitro by human blood outgrowth endothelial cells relative to vessel-derived endothelial cells. Tissue Engineering. 2005;11:1332–1345. doi: 10.1089/ten.2005.11.1332. [DOI] [PubMed] [Google Scholar]
- [194].Ramanujan S, Pluen A, McKee TD, Brown EB, Boucher Y, Jain RK. Diffusion and convection in collagen gels: Implications for transport in the tumor interstitium. Biophysical Journal. 2002;83:1650–1660. doi: 10.1016/S0006-3495(02)73933-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Cross VL, Zheng Y, Choi NW, Verbridge SS, Sutermaster BA, Bonassar LJ, Fischbach C, Stroock AD. Dense type I collagen matrices that support cellular remodeling and microfabrication for studies of tumor angiogenesis and vasculogenesis in vitro. Biomaterials. 2010;31:8596–8607. doi: 10.1016/j.biomaterials.2010.07.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Anderson ARA, Quaranta V. Integrative mathematical oncology. Nat. Rev. Cancer. 2008;8:227–234. doi: 10.1038/nrc2329. [DOI] [PubMed] [Google Scholar]
- [197].Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J, Cook K, Stepansky A, Levy D, Esposito D, Muthuswamy L, Krasnitz A, McCombie WR, Hicks J, Wigler M. Tumour evolution inferred by single-cell sequencing. Nature. 2011;472:90–U119. doi: 10.1038/nature09807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Campbell PJ, Pleasance ED, Stephens PJ, Dicks E, Rance R, Goodhead I, Follows GA, Green AR, Futreal PA, Stratton MR. Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:13081–13086. doi: 10.1073/pnas.0801523105. [DOI] [PMC free article] [PubMed] [Google Scholar]




