Skip to main content
BME Frontiers logoLink to BME Frontiers
. 2023 Jan 16;4:0006. doi: 10.34133/bmef.0006

Challenges and Opportunities Modeling the Dynamic Tumor Matrisome

Shelly R Peyton 1,*,, Manu O Platt 2,, Edna Cukierman 3,
PMCID: PMC10521682  PMID: 37849664

Abstract

We need novel strategies to target the complexity of cancer and, particularly, of metastatic disease. As an example of this complexity, certain tissues are particularly hospitable environments for metastases, whereas others do not contain fertile microenvironments to support cancer cell growth. Continuing evidence that the extracellular matrix (ECM) of tissues is one of a host of factors necessary to support cancer cell growth at both primary and secondary tissue sites is emerging. Research on cancer metastasis has largely been focused on the molecular adaptations of tumor cells in various cytokine and growth factor environments on 2-dimensional tissue culture polystyrene plates. Intravital imaging, conversely, has transformed our ability to watch, in real time, tumor cell invasion, intravasation, extravasation, and growth. Because the interstitial ECM that supports all cells in the tumor microenvironment changes over time scales outside the possible window of typical intravital imaging, bioengineers are continuously developing both simple and sophisticated in vitro controlled environments to study tumor (and other) cell interactions with this matrix. In this perspective, we focus on the cellular unit responsible for upholding the pathologic homeostasis of tumor-bearing organs, cancer-associated fibroblasts (CAFs), and their self-generated ECM. The latter, together with tumoral and other cell secreted factors, constitute the “tumor matrisome”. We share the challenges and opportunities for modeling this dynamic CAF/ECM unit, the tools and techniques available, and how the tumor matrisome is remodeled (e.g., via ECM proteases). We posit that increasing information on tumor matrisome dynamics may lead the field to alternative strategies for personalized medicine outside genomics.

The Grand Challenges in Studying Tumor Stroma

Fibroblastic cells and their self-generated extracellular matrix (ECM) constitute a physiologic functional unit [1]. This basic connective tissue unit sustains the homeostasis of healthy organs and is responsible for orchestrating local and systemic responses to temporary perturbances like wounding or acute infections. Similarly, tumor-educated cancer-associated fibroblasts (CAFs) and the ECM they synthesize constitute a perturbed functional unit that upholds cancer homeostasis (Fig. 1) [2].

Fig. 1.

Fig. 1.

Cancer-associated fibroblasts (CAFs) and their self-generated ECM constitute the desmoplastic functional unit responsible for upholding the cancerous tissue homeostasis. Created with BioRender.com

Cancer is one of the leading causes of deaths in the US [3]. The majority of nonhereditary carcinomas, like lung, pancreas, liver, colorectal, breast, prostate, and others, develop from epithelial cells that undergo a series of oncogenic mutations. Nonetheless, somatic mutations alone do not fully explain epithelial tumor development. This is probably because this type of tumor onset and its progressive development necessitates that the naturally tumor-suppressive stroma is altered in a way that ceases to restrain tumorigenesis [46]. Hence, the noted “full organ” involvement in cancer development suggests that the Somatic Mutation Theory [7,8], arguing in favor of oncogenic-driver mutations, and the Tissue Organization Field Theory [911], proposing that the stroma constitutes a natural tumor suppressor in need to be modulated to support tumor development, are mutually inclusive and can collectively explain the local and systemic aspects of tumorigenesis [4]. This idea can begin to explain the reason why numerous attempts to eliminate the stroma have provided no benefit to patients and occasionally have even been detrimental [12].

Importantly, the above-mentioned CAF/ECM functional unit is the main stroma component of solid epithelial cancers, known as desmoplasia [13]. Desmoplasia, described as a chronic healing-like fibroblastic stroma field expansion, favors tumorigenesis by nurturing tumor cells and concurrently sustaining pro-tumor immunosuppression [13]. Nonetheless, while normal stroma is naturally tumor-suppressive, tumor perturbed stroma is both tumor-supportive and tumor-suppressive [13]. Therefore, efforts to inhibit the former while harnessing the natural tumor-suppressive functions of desmoplasia justify ongoing preclinical efforts for attaining an effective “stroma normalization” that could be translated to the clinic.

To this end, since fibroblastic function and its modulation are dictated by the units’ ECM substrate, it is essential to realize that classic, 2-dimensional culturing of fibroblastic cells imposes an artificial type of stress upon these cells. The fibroblastic cell requirement of a pathophysiologic microenvironment for in vivo-like function substantiates efforts for culturing these cells within carefully selected matrix substrates, such as self-generated ECM [1]. Better understanding of the natural intact and cancer-perturbed interstitial ECMs should provide guidance for effective bioengineering of ECM-like substrates with high pathophysiologic relevance [1416]. Furthermore, studying how intact ECMs are altered to become perturbed ECMs that sustain cancer homeostasis is also of paramount importance [1719].

Knowing the central role that fibroblastic cells play in wound healing and understanding that sustained healing biases cancer development [20], it is posited that chronic assaults, such as frequent smoking, stress, obesity, “inflamaging,” and others, alter the fibroblastic cell/ECM in a way that this functional unit ceases to actively suppress tumor development. The possibility to detect tumor-predisposing traits and functions [13,2124] within this functional unit constitutes a highly sought out endeavor. Since the fibroblastic/ECM functional unit can be modulated by cell-intrinsic and -extrinsic factors, better understanding of this unit is necessary to design how stroma normalization could be attained.

Were all fibroblasts and all matrices in tumors the same, this problem would be easier to solve and to translate across different types of patients. However, cancer is not a disease of conformity. Heterogeneity exists across different patients, from one cancer type to another, within the same cancer dispersed at different organs, and even across the cells within a single tumor. This heterogeneity complicates therapeutic strategies, obviously limits the benefits of targeting drugs to a dominant cancer genotype [25], and likely hinders efficacy of immunotherapy [26]. When including the heterogeneity of the stroma [27], matrix [28], etc., a more complicated environment is considered and opens further experimental pathways to be investigated. Engineers have stepped in to develop systems to modulate the tumor ECM with biomaterials controlling architecture and mechanics as well as varying presentation of synthetic polymers, natural polymers, glycans, and soluble factors [29], which we will discuss here, particularly within the context of the dynamic, continually remodeled CAF/ECM unit.

Remodeling of the Tumor Stroma

The stroma of solid epithelial cancers, including its scaffolding interstitial ECM, generated and sustained by a reciprocal crosstalk with local CAFs (e.g., CAF/ECM units), constitutes a key feature of tumorigenesis. This is because the tumor-associated stroma can generate barriers and paths, which cage or gate the entrance as well as the escape of key cellular components (e.g., cancer cell metastatic escape or immune cell access). Being both physical and biochemical, these barriers and paths modulate the location as well as the functional states of a plethora of cancer and tumor microenvironment (TME) cells. An important feature of the above-mentioned pathologic homeostasis is explained through the clear increase in tumor and stroma secreted TME modulators. For example, high levels of cysteine cathepsins, matrix metalloproteases, and other proteases [3036] specialize on cleaving unique components of the epithelial and the interstitial ECMs such as laminin, collagens type I and IV, and fibronectin [3739]. Proteases can cleave this ECM to make large spaces and overcome physical barriers, thus facilitating the metastatic escape of cancer cells in bulk or as single cells (e.g., through an epithelial-to-mesenchymal transition). Importantly, if the generated gaps do not render sufficient access, cancer cells can still escape by altering their migratory strategy, using a mesenchymal-to-ameboid switch, and squeeze through the protease-generated and natural ECM pores [40].

Of note, dynamic interstitial ECM changes are a noteworthy requirement of in vivo tumorigenesis. To this end, the late Dr. Keely revolutionized the biophysics interstitial ECM field in a series of seminal discoveries. The first demonstrated that the biophysical state of fibrous, collagen-I-rich ECMs (e.g., connective tissue interstitial ECM) dictates epithelial cell function [41,42]. The second revealed that during tumorigenesis, fibroblast/ECM units undergo a dynamic reorganization of collagen I ECM fibrils, which result in the formation of 3 possible types of distinct and measurable “tumor-associated collagen signatures”, known as TACS-1, TACS-2, and TACS-3 [43]. Importantly, Keely (and others after her) demonstrated that patients with surgically resected tumors that encompass high levels of TACS-3 (e.g., ECM fibers that are parallely aligned and oriented perpendicularly to the tumor border) within the TME experience shorter overall survival [4447]. This increase in the amount and alignment of collagen fibers also stiffens the ECM at the tumor border, from hundreds of pascals to tens or even hundreds of kilopascals [43,4860].

Furthermore, individual matrix proteins have been identified that either suppress overt metastasis, including thrombospondin 1 on the microvascular endothelium [61], or facilitate it, such as in the case of CAF/ECM units generating differentially spliced variants of fibronectin [6264]. Moreover, tumor cells can trigger stromal periostin secretion to metastasize to the lung [65], and breast cancer cell-derived tenascin C promotes lung metastasis [66]. As other examples, ECM proteins like fibronectin, collagen, and others, as well as their protease-generated fragments, can support the survival of incoming disseminated tumor cells [67,68]. Notably, some of the proteins, including many key chemokines and cytokines that signal to local cancer and stroma cells, are activated or facilitated by protease activity; many are the result of ECM fragmentation, while others are released and often even activated due to ECM cleavage [69] (Fig. 2).

Fig. 2.

Fig. 2.

Cathepsins and other proteases released by CAFs and TAMs mediate degradation of ECM at the tumor boundary, leading to cancer cell escape to nearby blood vessels and eventual metastases. Created with BioRender.com

Beyond the tumor cells and the CAF/ECM units, another central player promoting cancer cell escape from the primary tumor site is the tumor-associated macrophage (TAM) [70]. Infiltration of TAMs is associated with poor prognosis [7175], and at the tumor, TAMs promote angiogenesis [74,7680], chemotherapy resistance [81], tumor growth [76], invasion, metastasis [82], and tumor-promoting immune responses [8386], and secrete tissue-remodeling cysteine cathepsin proteases [8790] and other proteases [9193] (Fig. 2). TAM-secreted proteases play a critical role at the primary tumor [81,87]. This could implicate targeting of macrophages, or even monocytes, in the marrow milieu as a powerful mechanism to decrease metastatic burden. Studies have also shown that monocyte-derived macrophages as models for TAMs can assist cancer cell invasion in patient-specific ways [94,95].

Experimentally Modeling the Dynamic TME

Bioengineers are continuously iterating on and refining the experimental systems in which to study the impact of the CAF/ECM unit on tumor growth, progression, metastasis, and drug response, as well as how the tumor matrisome co-evolves with the tumor cells, via remodeling or otherwise. Thus far, the gold standard has been rodent models. The field acknowledges that rodent models do not perfectly reflect human disease [96], yet it is currently not possible to validate findings from in vitro experiments any other way. Rodent models often lack a human-like stromal architecture, have a rate of disease progression that far outpaces that in humans, and do not have a human immune system (some models have a near lack of any immune system) [97].

To bridge the gap between oversimplified plastic dishes and rodent models, bioengineers have been creating increasingly more complex and realistic environments to mimic the human TME. The NCI, originally partnering with the NSF, has established venues to specifically fund this work through the Physical and Engineering Sciences in Oncology program (NSF 12-514). Phenomenal advances have been made in tissue engineering to create both organs and systems of tissues [98,99]. By eventually combining these with tumor studies, the dream is that it may be possible to replace rodent models with these engineered systems and do all cancer work in an entirely human-like dynamic TME system.

Toward these more human-like studies, there is increasing effort to use patient-derived tumoroids, organoids, and ex vivo tumor slices in in vitro cancer research, as they better reflect the genetic makeup of tumors in patients. This field is new, but far more is known about the genetic makeup and selection of cells during organoid culture compared to the makeup of the stroma in these patient-derived organoids. Pioneering work from the Hynes lab has defined the matrisome, including the insoluble scaffolding ECM and other secreted factors, via semiquantitative mass spectrometry and provided substantial detail on the matrisomal stroma of mouse tumors [100], which has been corroborated with gene expression of stroma markers in humans [101,102]. To date, we do not know the composition of the matrisome of patient-derived organoids in in vitro systems, and this will be key to determining if these tumor models are reflective of the real TME. Patient-derived organoids are also an opportunity to better understand sex- and ancestry-driven differences in TME dynamics, which has thus far not been addressed. Given the emerging information on sex- and ancestry-driven differences in cell sourcing and disease [103105], this is a critical gap in matrisome knowledge. Yet, organoids do not often sustain/include stromal cells and thus their TME remains limited.

Perturbing the matrisome of tumors in vivo is challenging. Many ECM protein deletions are embryonically lethal, and conditional mouse models are not economically or technologically accessible to most laboratories (in the US, let alone globally). The ability to perturb the matrisome on the bench is incredibly valuable for hypothesis testing before moving to preclinical models. Several model systems, in vitro, exist where one could replicate many components of the matrisome and include cancer cells or organoids. Matrigel, despite its drawbacks, remains the most widely used of these in vitro cancer cell culture environments [106]. It is very easy to use and contains nearly all the complexity of the basement membrane, which constitutes an important aspect of the TME matrisome of many solid cancers. The overwhelming frustration with Matrigel is its variability, a weariness that is exaggerated given the high cost. Furthermore, Matrigel in the normal basement membrane does not facilitate invasion, but instead is a barrier for solid cancer invasion. Matrigel contains the proteins from the basement membrane (and many more), but it is typically reconstituted in vitro in a manner not tightly crosslinked like the native basement membrane. The native epithelial basement membrane sustains cancer cell growth and simultaneously provides a barrier for cancer cell invasion. In contrast, the interstitial matrix sustained in vivo by CAF/ECM units that is needed to facilitate metastatic escape is often modeled using fibrillar collagen type 1 gels.

However, other systems, despite development effort across many groups, still fall short of Matrigel and other cell-derived ECMs’ performances. Then, again, polymeric hydrogels are advantageous because they are immensely controllable—the end user can carefully dial-in a desired modulus, integrin-binding proteins or peptides included [14,16], tailor the degradation of the cell-encapsulating material, and control the porosity using leaching techniques or allow cells to move by including protease-cleavable crosslinks [107]. Drawbacks to these hydrogels are that they do not have the 3-dimensional structure of native basement membranes or the 3-dimensional fibrillar topography of interstitial ECMs (e.g., generated naturally by tumor-educated CAFs), the posttranslational modifications on native proteins, or the richness of growth-factor-binding sites typical of sugars and proteoglycans offered by native matrices [108]. Peptide-based hydrogels are controllable and give a fibrillar structure akin to the interstitial ECM (reviewed in [109]). Cell attachment points rely on serum and cell-secreted proteins binding electrostatically to the fibrillar matrices or by incorporating integrin-binding peptide fragments into the sequence. Their use as cancer models has been limited, but if one could build more functionality into the peptides [14], or control secreted proteins to represent that of the CAF/ECM unit, these could become powerful models. Cell-derived ECMs are one of the best alternatives, as they have the natural protein structure, yet they are only tunable if one can control the fibroblastic ECM-secreting and -organizing proteins in culture, as well as the underlying stiffness of the substrate used to attain cell-derived ECM fibrillogenesis [24,110,111]. For example, fibroblasts can be isolated from healthy tissue, tumor-adjacent tissue, or intratumoral CAFs (e.g., educated by the tumor; Fig. 1). Fibroblasts can also be biomechanically, genetically, and/or biochemically modulated (e.g., using defined mixtures of growth factors and cytokines), and thus engineered to produce and organize specific types of ECM presenting altered functions [22,45,110112].

The immune component of the TME, which is not a focus of this particular perspective, has been studied in vivo, but in vitro experiments are far behind. Macrophages, NK cells, neutrophils, and cytotoxic T cells, all important for tumor cell killing in immunotherapies, are short lived in vivo, and strategies to keep them alive for study in vitro are in development. Ideally, one could study the evolution of immune environment alongside the stroma in vitro to hypothesis test and screen immunotherapies. A major roadblock remains, however, in that it is technically challenging to culture most immune cells (macrophages are an exception) in in vitro cell culture environments. Recent advancements have included those from patient-derived organoids, which include all components of the tumor all at once rather than combining isolated components from different sources (for a more complete review on these systems, see [113]).

Computational and mathematical models can be helpful to integrate these complexities: agent-based models, mathematical models of mechanics incorporating biophysical parameters, analytical models of stochastic dynamics of mutations, and kinetic equations incorporating cell growth and death are examples used to assess and derive predictions of both collective and individual tumor cell responses. Systems biology approaches are used to analyze, integrate, and bridge data derived from in vitro and in vivo approaches [114118] and simulations of CAF/ECM evolution alongside tumorigenesis [111,119,120]. Feedback loops elicited by small-molecule therapies can, at times, be developed or predicted through merging experimental and computational approaches [17,19,25]. Furthermore, computational models have been able to explain that to account for cell–ECM reciprocal interactions, the interstitial ECM necessitates specific viscoelastic properties that could have not been explained with the previous thinking of these scaffolds as plastic microenvironments [119]. Limitations of computational models in the data are collected to build the model and the correctness of assumptions used in their derivation, but their power lies in the manipulations and simulations that can be simulated to conserve time, reagents, and money incorporating physical, cellular, and environmental parameters.

Outlook

The ECM of the TME can confer unique survival advantages to cancer cells in the presence of otherwise powerful chemotherapies and targeted drugs. There is particular interest in targeting tumor desmoplasia as a way to kill cancer cells, in addition to, or as an alternative to personalized therapies targeting genetic mutations. First described for hypoxia [121], synthetic lethalities designed to hit the tumorigenic ECM are of increasing interest (reviewed in [122]). An overwhelming need is to study the intersection of CAF/ECM evolution and immunotherapy to increase T-cell infiltration into tumors and potentially understand how the ECM regulates T-cell activation and exhaustion.

It is not yet clear if the concept of genetic (including epigenetic) heterogeneity across tumors, organs, and individuals extends to the ECM. Matrisome studies, in fact, suggest that the desmoplastic ECM presents with commonalities across patients and tumor types. We hence wonder if potentially identifying unifying themes among types of tumors despite their organ location could render means to regulate and/or prevent tumorigenesis. Then, again, the fact that certain chronic conditions can solely predispose patients to develop specific cancers, like in the case of pancreatic cancer linked to chronic pancreatitis, suggests that key differences in stroma could encourage unique tumor onsets. Could this type of particular and common tumor stroma-predisposing information help oncologists reach a broader patient population? After all, themes around the CAF/ECM unit that are ubiquitous (or indicative of predispositions) could assist in reducing patient pools based solely on ultra-targeted, and often very expensive, genetic-motivated therapies. Focusing on the TME (including CAF/ECM functional units) could get us to identify “generic” or at least more inclusive and powerful cross-cutting tools.

Acknowledgments

We dedicate this work to Patricia Keely, a role model scientist and pioneer in the field of interstitial ECM biology.

Funding: This work was supported by a grant from the Jayne Koskinas Ted Giovanis Foundation for Health and Policy and a grant from the National Institutes of Health (NIH) (U01CA265709) to S.R.P. S.R.P. was supported by an Armstrong Professorship at UMass Amherst. M.O.P. was supported by a donation from the Giglio Family to the Wallace H. Coulter Department of Biomedical Engineering. E.C. was supported by funds from the Pancreatic Cancer Cure Foundation, and the Concetta Greenberg Pancreatic Cancer Institute. Grants to E.C. that supported this work were from the Pennsylvania’s DOH Health Research Formula Funds, the 5th AHEPA Cancer Research Foundation, Inc., the Worldwide Cancer Research, and the NIH/NCI (R21CA231252, R21CA252535, R01CA232256, R01CA269660, S10ODO23666, and P30CA06927).

Author contributions: S.R.P., M.O.P., and E.C. equally contributed to the writing of this manuscript.

Competing interests: E.C. is a consultant in oncologic stroma biology for Phenomic AI and Takeda.

References

  • 1.Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix adhesions to the third dimension. Science. 2001;294(5547):1708–1712. [DOI] [PubMed] [Google Scholar]
  • 2.Gardiner JC, Cukierman E. Meaningful connections: Interrogating the role of physical fibroblast cell–cell communication in cancer. In: Angel P, Ostrowski M, editors. Stromal signaling in cancer. Advances in cancer research. 1st ed. Elsevier; Vol. 154, 2022. p. 1–251. [DOI] [PMC free article] [PubMed]
  • 3.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. [DOI] [PubMed] [Google Scholar]
  • 4.Alexander J, Cukierman E. Cancer associated fibroblast: Mediators of tumorigenesis. Matrix Biol. 2020;91–92:19–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Illmensee K, Mintz B. Totipotency and normal differentiation of single teratocarcinoma cells cloned by injection into blastocysts. Proc Natl Acad Sci USA. 1976;73(2):549–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mintz B, Illmensee K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci USA. 1975;72(9):3585–3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Boveri T. Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J Cell Sci. 2008;121(Suppl. 1):1–84. [DOI] [PubMed] [Google Scholar]
  • 8.Boveri T. Anton Dohbn. Science. 1912;36(928):453–468. [DOI] [PubMed] [Google Scholar]
  • 9.Soto AM, Longo G, Montevil M, Sonnenschein C. The biological default state of cell proliferation with variation and motility, a fundamental principle for a theory of organisms. Prog Biophys Mol Biol. 2016;122(1):16–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sonnenschein C, Soto AM. Theories of carcinogenesis: An emerging perspective. Semin Cancer Biol. 2008;18(5):372–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Triolo VA. Nineteenth century foundations of cancer research. Orig Exp Res Cancer Res. 1964;24:4–27. [PubMed] [Google Scholar]
  • 12.Kim EJ, Sahai V, Abel EV, Griffith KA, Greenson JK, Takebe N, Khan GN, Blau JL, Craig R, Balis UG, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin Cancer Res. 2014;20(23):5937–5945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20(3):174–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jansen LE, Kim H, Hall CL, McCarthy TP, Lee MJ, Peyton SR. A poly(ethylene glycol) three-dimensional bone marrow hydrogel. Biomaterials. 2022;280:121270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barney LE, Dandley EC, Jansen LE, Reich NG, Mercurio AM, Peyton SR. A cell-ECM screening method to predict breast cancer metastasis. Integrative biology: Quantitative biosciences from nano to macro. 2015;7(2):198–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Galarza S, Crosby AJ, Pak C, Peyton SR. Control of astrocyte quiescence and activation in a synthetic brain hydrogel. Adv Healthc Mater. 2020;9(4):e1901419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wilder CL, Walton C, Watson V, Stewart FAA, Johnson J, Peyton SR, Payne CK, Odero-Marah V, Platt MO. Differential cathepsin responses to inhibitor-induced feedback: E-64 and cystatin C elevate active cathepsin S and suppress active cathepsin L in breast cancer cells. Int J Biochem Cell Biol. 2016;79:199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Barney LE, Hall CL, Schwartz AD, Parks AN, Sparages C, Galarza S, Platt MO, Mercurio AM, Peyton SR. Tumor cell-organized fibronectin maintenance of a dormant breast cancer population. Sci Adv. 2020;6(11):eaaz4157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shockey WA, Kieslich CA, Wilder CL, Watson V, Platt MO. Dynamic model of protease state and inhibitor trafficking to predict protease activity in breast cancer cells. Cell Mol Bioeng. 2019;12(4):275–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dvorak HF. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–1659. [DOI] [PubMed] [Google Scholar]
  • 21.Gardiner JC, Raghavan KS, Alexander JI, Franco-Barraza J, Cukierman E. CHAPTER 16. Fibroblastic cell-derived extracellular matrices: A cell culturing system to model key aspects of the tumor microenvironment. Decellularized extracellular matrix. Biomaterials Sci Ser 6 R Soc Chem 2020. 2019;6:305–327. [Google Scholar]
  • 22.Francescone R, Barbosa Vendramini-Costa D, Franco-Barraza J, Wagner J, Muir A, Lau AN, Gabitova L, Pazina T, Gupta S, Luong T, et al. Netrin G1 promotes pancreatic tumorigenesis through cancer associated fibroblast-driven nutritional support and immunosuppression. Cancer Discov. 2021;11(2):446–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Franco-Barraza J, Raghavan KS, Luong T, Cukierman E. Engineering clinically-relevant human fibroblastic cell-derived extracellular matrices. Methods Cell Biol. 2020;156:109–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Franco-Barraza J, Francescone R, Luong T, Shah N, Madhani R, Cukierman G, Dulaimi E, Devarajan K, Egleston BL, Nicolas E, et al. Matrix-regulated integrin αvβ5 maintains α5β1-dependent desmoplastic traits prognostic of neoplastic recurrence. eLife. 2017;6:e20600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhao B, Hemann MT, Lauffenburger DA. Intratumor heterogeneity alters most effective drugs in designed combinations. Proc Natl Acad Sci USA. 2014;111(29):10773–10778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vitale I, Shema E, Loi S, Galluzzi L. Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med. 2021;27(2):212–224. [DOI] [PubMed] [Google Scholar]
  • 27.Landry BD, Leete T, Richards R, Cruz-Gordillo P, Schwartz HR, Honeywell ME, Ren G, Schwartz AD, Peyton SR, Lee MJ. Tumor-stroma interactions differentially alter drug sensitivity based on the origin of stromal cells. Mol Syst Biol. 2018;14(8):e8322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pally D, Goutham S, Bhat R. Extracellular matrix as a driver for intratumoral heterogeneity. Phys Biol. 2022;19(4):043001. [DOI] [PubMed] [Google Scholar]
  • 29.Shimpi AA, Fischbach C. Engineered ECM models: Opportunities to advance understanding of tumor heterogeneity. Curr Opin Cell Biol. 2021;72:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chauhan SS, Goldstein LJ, Gottesman MM. Expression of cathepsin L in human tumors. Cancer Res. 1991;51(5):1478–1481. [PubMed] [Google Scholar]
  • 31.Zhang J, Shridhar R, Dai Q, Song J, Barlow SC, Yin L, Sloane BF, Miller FR, Meschonat C, Li BDL, et al. Cystatin m: A novel candidate tumor suppressor gene for breast cancer. Cancer Res. 2004;64(19):6957–6964. [DOI] [PubMed] [Google Scholar]
  • 32.Kopitz C, Anton M, Gansbacher B, Kruger A. Reduction of experimental human fibrosarcoma lung metastasis in mice by adenovirus-mediated cystatin C overexpression in the host. Cancer Res. 2005;65(19):8608–8612. [DOI] [PubMed] [Google Scholar]
  • 33.Li W, Ding F, Zhang L, Liu Z, Wu Y, Luo A, Wu M, Wang M, Zhan Q, Liu Z. Overexpression of stefin A in human esophageal squamous cell carcinoma cells inhibits tumor cell growth, angiogenesis, invasion, and metastasis. Clin Cancer Res. 2005;11(24 Pt 1):8753–8762. [DOI] [PubMed] [Google Scholar]
  • 34.Podgorski I, Linebaugh BE, Koblinski JE, Rudy DL, Herroon MK, Olive MB, Sloane BF. Bone marrow-derived cathepsin K cleaves SPARC in bone metastasis. Am J Pathol. 2009;175(3):1255–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mohamed MM, Sloane BF. Cysteine cathepsins: Multifunctional enzymes in cancer. Nat Rev Cancer. 2006;6(10):764–775. [DOI] [PubMed] [Google Scholar]
  • 36.Chen B, Platt MO. Multiplex zymography captures stage-specific activity profiles of cathepsins K, L, and S in human breast, lung, and cervical cancer. J Transl Med. 2011;9:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Buck MR, Karustis DG, Day NA, Honn KV, Sloane BF. Degradation of extracellular-matrix proteins by human cathepsin B from normal and tumour tissues. Biochem J. 1992;282(Pt. 1):273–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Guinec N, Dalet-Fumeron V, Pagano M. "In vitro" study of basement membrane degradation by the cysteine proteinases, cathepsins B, B-like and L. Digestion of collagen IV, laminin, fibronectin, and release of gelatinase activities from basement membrane fibronectin. Biol Chem Hoppe Seyler. 1993;374(12):1135–1146. [DOI] [PubMed] [Google Scholar]
  • 39.Ishidoh K, Kominami E. Procathepsin L degrades extracellular matrix proteins in the presence of glycosaminoglycans in vitro. Biochem Biophys Res Commun. 1995;217(2):624–631. [DOI] [PubMed] [Google Scholar]
  • 40.Wolf K, Mazo I, Leung H, Engelke K, Andrian UH, Deryugina EI, Strongin AY, Bröcker EB, Friedl P. Compensation mechanism in tumor cell migration: Mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 2003;160(2):267–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wozniak MA, Desai R, Solski PA, Der CJ, Keely PJ. ROCK-generated contractility regulates breast epithelial cell differentiation in response to the physical properties of a three-dimensional collagen matrix. J Cell Biol. 2003;163(3):583–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.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 Med. 2008;6:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med. 2006;4(1):38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, Friedl A, Keely PJ. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol. 2011;178(3):1221–1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Goetz JG, Minguet S, Navarro-Lerida I, Lazcano JJ, Samaniego R, Calvo E, Tello M, Osteso-Ibáñez T, Pellinen T, Echarri A, et al. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell. 2011;146(1):148–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Conklin MW, Gangnon RE, Sprague BL, Van Gemert L, Hampton JM, Eliceiri KW, Bredfeldt JS, Liu Y, Surachaicharn N, Newcomb PA, et al. Collagen alignment as a predictor of recurrence after ductal carcinoma in situ. Cancer Epidemiol Biomarkers Prev. 2018;27(2):138–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Drifka CR, Loeffler AG, Mathewson K, Keikhosravi A, Eickhoff JC, Liu Y, Weber SM, Kao WJ, Eliceiri KW. Highly aligned stromal collagen is a negative prognostic factor following pancreatic ductal adenocarcinoma resection. Oncotarget. 2016;7(46):76197–76213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A, Weninger W, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer. Dis Model Mech. 2011;4(2):165–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, Chen YY, Liphardt J, Hwang ES, Weaver VM. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol. 2015;7(10):1120–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Katira P, Bonnecaze RT, Zaman MH. Modeling the mechanics of cancer: Effect of changes in cellular and extra-cellular mechanical properties. Front Oncol. 2013;3:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Seo BR, Bhardwaj P, Choi S, Gonzalez J, Eguiluz RCA, Wang K, Mohanan S, Morris PG, Du B, Zhou XK, et al. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci Trans Med. 2015;7(301):301ra130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Huwart L, Salameh N, Beek L, Vicaut E, Peeters F, Sinkus R, van Beers BE. MR elastography of liver fibrosis: Preliminary results comparing spin-echo and echo-planar imaging. Eur Radiol. 2008;18(11):2535–2541. [DOI] [PubMed] [Google Scholar]
  • 54.Huwart L, Peeters F, Sinkus R, Annet L, Salameh N, Beek LC, Horsmans Y, van Beers BE. Liver fibrosis: Non-invasive assessment with MR elastography. NMR Biomed. 2006;19(2):173–179. [DOI] [PubMed] [Google Scholar]
  • 55.Tilghman RW, Cowan CR, Mih JD, Koryakina Y, Gioeli D, Slack-Davis JK, Blackman BR, Tschumperlin DJ, Parsons JT. Matrix rigidity regulates cancer cell growth and cellular phenotype. PLOS ONE. 2010;5(9):e12905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8(3):241–254. [DOI] [PubMed] [Google Scholar]
  • 57.Paszek MJ, Weaver VM. The tension mounts: Mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia. 2004;9(4):325–342. [DOI] [PubMed] [Google Scholar]
  • 58.Bretscher MS. On the shape of migrating cells—A ‘front-to-back’ model. J Cell Sci. 2008;121(Pt. 16):2625–2628. [DOI] [PubMed] [Google Scholar]
  • 59.Iwanicki MP, Davidowitz RA, Ng MR, Besser A, Muranen T, Merritt M, Danuser G, Ince TA, Brugge JS. Ovarian cancer spheroids use myosin-generated force to clear the mesothelium. Cancer Discov. 2011;1(2):144–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kostic A, Lynch CD, Sheetz MP. Differential matrix rigidity response in breast cancer cell lines correlates with the tissue tropism. PLOS ONE. 2009;4(7):e6361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, Almeida D, Koller A, Hajjar KA, Stainier DYR, et al. The perivascular niche regulates breast tumour dormancy. Nat Cell Biol. 2013;15(7):807–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sun X, Fa P, Cui Z, Xia Y, Sun L, Li Z, Tang A, Gui Y, Cai Z. The EDA-containing cellular fibronectin induces epithelial-mesenchymal transition in lung cancer cells through integrin α9β1-mediated activation of PI3-K/AKT and Erk1/2. Carcinogenesis. 2014;35(1):184–191. [DOI] [PubMed] [Google Scholar]
  • 63.Kunz-Schughart LA, Knuechel R. Tumor-associated fibroblasts (Part I): Active stromal participants in tumor development and progression? Histol Histopathol. 2002;17(2):599–621. [DOI] [PubMed] [Google Scholar]
  • 64.Schor SL. Fibroblast subpopulations as accelerators of tumor progression: The role of migration stimulating factor. Exs. 1995;74:273–296. [DOI] [PubMed] [Google Scholar]
  • 65.Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HA, Delaloye JF, Huelsken J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011;481(7379):85–89. [DOI] [PubMed] [Google Scholar]
  • 66.Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, Downey RJ, Manova-Todorova K, Brogi E, et al. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat Med. 2011;17(7):867–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kaplan RN, Psaila B, Lyden D. Bone marrow cells in the 'pre-metastatic niche': Within bone and beyond. Cancer Metastasis Rev. 2006;25(4):521–529. [DOI] [PubMed] [Google Scholar]
  • 68.Sanford-Crane H, Abrego J, Sherman MH. Fibroblasts as modulators of local and systemic cancer metabolism. Cancers. 2019;11(5):619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cox TR. The matrix in cancer. Nat Rev Cancer. 2021;21(4):217–238. [DOI] [PubMed] [Google Scholar]
  • 70.Goswami S, Sahai E, Wyckoff JB, Cammer M, Cox D, Pixley FJ, Stanley ER, Segall JE, Condeelis JS. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005;65(12):5278–5283. [DOI] [PubMed] [Google Scholar]
  • 71.DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1(1):54–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Laoui D, Movahedi K, Van Overmeire E, Van den Bossche J, Schouppe E, Mommer C, Nikolaou A, Morias Y, De Baetselier P, Van Ginderachter JA. Tumor-associated macrophages in breast cancer: Distinct subsets, distinct functions. Int J Dev Biol. 2011;55(7-9):861–867. [DOI] [PubMed] [Google Scholar]
  • 73.Lee AH, Happerfield LC, Bobrow LG, Millis RR. Angiogenesis and inflammation in invasive carcinoma of the breast. J Clin Pathol. 1997;50(8):669–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL. Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 1996;56(20):4625–4629. [PubMed] [Google Scholar]
  • 75.Campbell MJ, Tonlaar NY, Garwood ER, Huo D, Moore DH, Khramtsov AI, Au A, Baehner F, Chen Y, Malaka DO, et al. Proliferating macrophages associated with high grade, hormone receptor negative breast cancer and poor clinical outcome. Breast Cancer Res Treat. 2011;128(3):703–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–612. [DOI] [PubMed] [Google Scholar]
  • 77.Lewis JS, Landers RJ, Underwood JC, Harris AL, Lewis CE. Expression of vascular endothelial growth factor by macrophages is up-regulated in poorly vascularized areas of breast carcinomas. J Pathol. 2000;192(2):150–158. [DOI] [PubMed] [Google Scholar]
  • 78.Dirkx AE, Oude Egbrink MGA, Wagstaff J, Griffioen AW. Monocyte/macrophage infiltration in tumors: Modulators of angiogenesis. J Leukoc Biol. 2006;80(6):1183–1196. [DOI] [PubMed] [Google Scholar]
  • 79.Roland CL, Dineen SP, Lynn KD, Sullivan LA, Dellinger MT, Sadegh L, Sullivan JP, Shames DS, Brekken RA. Inhibition of vascular endothelial growth factor reduces angiogenesis and modulates immune cell infiltration of orthotopic breast cancer xenografts. Mol Cancer Ther. 2009;8(7):1761–1771. [DOI] [PubMed] [Google Scholar]
  • 80.Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP. Amino-biphosphonate-mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res. 2007;67(23):11438–11446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Shree T, Olson OC, Elie BT, Kester JC, Garfall AL, Simpson K, Bell-McGuinn KM, Zabor EC, Brogi E, Joyce JA. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 2011;25(23):2465–2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med. 2001;193(6):727–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Balkwill F, Mantovani A. Inflammation and cancer: Back to Virchow? Lancet. 2001;357(9255):539–545. [DOI] [PubMed] [Google Scholar]
  • 84.Sica A, Schioppa T, Mantovani A, Allavena P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. Eur. J. Cancer. 2006;42(6):717–727. [DOI] [PubMed] [Google Scholar]
  • 85.Mantovani A. B cells and macrophages in cancer: Yin and yang. Nat Med. 2011;17(3):285–286. [DOI] [PubMed] [Google Scholar]
  • 86.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–444. [DOI] [PubMed] [Google Scholar]
  • 87.Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T, Joyce JA. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010;24(3):241–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gocheva V, Zeng W, Ke D, Klimstra D, Reinheckel T, Peters C, Hanahan D, Joyce JA. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev. 2006;20(5):543–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Vasiljeva O, Papazoglou A, Kruger A, Brodoefel H, Korovin M, Deussing J, Augustin N, Nielsen BS, Almholt K, Bogyo M, et al. Tumor cell-derived and macrophage-derived cathepsin B promotes progression and lung metastasis of mammary cancer. Cancer Res. 2006;66(10):5242–5250. [DOI] [PubMed] [Google Scholar]
  • 90.Roodman GD. Mechanisms of bone metastasis. N Engl J Med. 2004;350(16):1655–1664. [DOI] [PubMed] [Google Scholar]
  • 91.Coulson-Thomas VJ, Gesteira TF, Coulson-Thomas YM, Vicente CM, Tersariol IL, Nader HB, Toma L. Fibroblast and prostate tumor cell cross-talk: Fibroblast differentiation, TGF-beta, and extracellular matrix down-regulation. Exp Cell Res. 2010;316(19):3207–3226. [DOI] [PubMed] [Google Scholar]
  • 92.Rasmussen HS, McCann PP. Matrix metalloproteinase inhibition as a novel anticancer strategy: A review with special focus on batimastat and marimastat. Pharmacol Ther. 1997;75(1):69–75. [DOI] [PubMed] [Google Scholar]
  • 93.Sabeh F, Li XY, Saunders TL, Rowe RG, Weiss SJ. Secreted versus membrane-anchored collagenases: Relative roles in fibroblast-dependent collagenolysis and invasion. J Biol Chem. 2009;284(34):23001–23011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Park KY, Li WA, Platt MO. Patient specific proteolytic activity of monocyte-derived macrophages and osteoclasts predicted with temporal kinase activation states during differentiation. Integr Biol. 2012;4(12):1459–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Park KY, Li G, Platt MO. Monocyte-derived macrophage assisted breast cancer cell invasion as a personalized, predictive metric to score metastatic risk. Sci Rep. 2015;5:13855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ben-David U, Ha G, Tseng Y-Y, Greenwald NF, Oh C, Shih J, McFarland JM, Wong B, Boehm JS, Beroukhim R, et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet. 2017;49(11):1567–1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Guerin MV, Finisguerra V, Van den Eynde BJ, Bercovici N, Trautmann A. Preclinical murine tumor models: A structural and functional perspective. eLife. 2020;9:e50740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ronaldson-Bouchard K, Teles D, Yeager K, Tavakol DN, Zhao Y, Chramiec A, Tagore S, Summers M, Stylianos S, Tamargo M, et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng. 2022;6(4):351–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Trapecar M, Wogram E, Svoboda D, Communal C, Omer A, Lungjangwa T, Sphabmixay P, Velazquez J, Schneider K, Wright CW, et al. Human physiomimetic model integrating microphysiological systems of the gut, liver, and brain for studies of neurodegenerative diseases. Sci Adv. 2021;7(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hebert JD, Myers SA, Naba A, Abbruzzese G, Lamar JM, Carr SA, Hynes RO. Proteomic profiling of the ECM of xenograft breast cancer metastases in different organs reveals distinct metastatic niches. Cancer Res. 2020;80(7):1475–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gocheva V, Naba A, Bhutkar A, Guardia T, Miller KM, Li CM, Dayton TL, Sanchez-Rivera FJ, Kim-Kiselak C, Jailkhani N, et al. Quantitative proteomics identify Tenascin-C as a promoter of lung cancer progression and contributor to a signature prognostic of patient survival. Proc Natl Acad Sci USA. 2017;114(28):E5625–E5634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Glavey SV, Naba A, Manier S, Clauser K, Tahri S, Park J, Reagan MR, Moschetta M, Mishima Y, Gambella M, et al. Proteomic characterization of human multiple myeloma bone marrow extracellular matrix. Leukemia. 2017;31(11):2426–2434. [DOI] [PubMed] [Google Scholar]
  • 103.Clarke S, Chin SN, Dodds L, George SHL, Badal S. Racial disparities in breast cancer preclinical and clinical models. Breast Cancer Res. 2022;24(1):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Raghavan S. How inclusive are cell lines in preclinical engineered cancer models? Dis Model Mech. 2022;15(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ryan H, Bister D, Holliday SA, Boehlein J, Lewis A, Silberman J, Allen JB, Moore E. Ancestral background is underreported in regenerative engineering. Regen Eng Transl Med. 2022;8(4):499–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Benton G, Arnaoutova I, George J, Kleinman HK, Koblinski J. Matrigel: From discovery and ECM mimicry to assays and models for cancer research. Adv Drug Deliv Rev. 2014;79–80:3–18. [DOI] [PubMed] [Google Scholar]
  • 107.Shi J, Yu L, Ding J. PEG-based thermosensitive and biodegradable hydrogels. Acta Biomater. 2021;128:42–59. [DOI] [PubMed] [Google Scholar]
  • 108.Goodarzi K, Rao SS. Hyaluronic acid-based hydrogels to study cancer cell behaviors. J Mater Chem B. 2021;9(31):6103–6115. [DOI] [PubMed] [Google Scholar]
  • 109.Worthington P, Pochan DJ, Langhans SA. Peptide hydrogels—Versatile matrices for 3D cell culture in cancer medicine. Front Oncol. 2015;5:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lee H-O, Mullins SR, Franco-Barraza J, Valianou M, Cukierman E, Cheng JD. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer. 2011;11(1):245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Malik R, Luong T, Cao X, Han B, Shah N, Franco-Barraza J, Han L, Shenoy VB, Lelkes PI, Cukierman E. Rigidity controls human desmoplastic matrix anisotropy to enable pancreatic cancer cell spread via extracellular signal-regulated kinase 2. Matrix Biol. 2019;81:50–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Alexander JI, Vendramini-Costa DB, Francescone R, Luong T, Franco-Barraza J, Shah N, Gardiner JC, Nicolas E, Raghavan KS, Cukierman E. Palladin isoforms 3 and 4 regulate cancer-associated fibroblast pro-tumor functions in pancreatic ductal adenocarcinoma. Sci Rep. 2021;11(1):3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Mengus C, Muraro MG, Mele V, Amicarella F, Manfredonia C, Foglietta F, Muenst S, Soysal SD, Iezzi G, Spagnoli GC. In vitro modeling of tumor-immune system interaction. ACS Biomater Sci Eng. 2018;4(2):314–323. [DOI] [PubMed] [Google Scholar]
  • 114.Leighow SM, Landry B, Lee MJ, Peyton SR, Pritchard JR. Agent-based models help interpret patterns of clinical drug resistance by contextualizing competition between distinct drug failure modes. bioRxiv. 2022;2022.02.25.481999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Leighow SM, Liu C, Inam H, Zhao B, Pritchard JR. Multi-scale predictions of drug resistance epidemiology identify design principles for rational drug design. Cell Rep. 2020;30(12):3951–3963.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bae SY, Guan N, Yan R, Warner K, Taylor SD, Meyer AS. Measurement and models accounting for cell death capture hidden variation in compound response. Cell Death Dis. 2020;11(4):255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Schwartz AD, Barney LE, Jansen LE, Nguyen TV, Hall CL, Meyer AS, Peyton SR. A biomaterial screening approach reveals microenvironmental mechanisms of drug resistance. Integr Biol. 2017;9(12):912–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schwartz HR, Richards R, Fontana RE, Joyce AJ, Honeywell ME, Lee MJ. Drug GRADE: An integrated analysis of population growth and cell death reveals drug-specific and cancer subtype-specific response profiles. Cell Rep. 2020;31(12):107800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chaudhuri O, Cooper-White J, Janmey PA, Mooney DJ, Shenoy VB. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature. 2020;584(7822):535–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Seo BR, Chen X, Ling L, Song YH, Shimpi AA, Choi S, Gonzalez J, Sapudom J, Wang K, Andresen Eguiluz RC, et al. Collagen microarchitecture mechanically controls myofibroblast differentiation. Proc Natl Acad Sci USA. 2020;117(21):11387–11398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Chan N, Pires IM, Bencokova Z, Coackley C, Luoto KR, Bhogal N, Lakshman M, Gottipati P, Oliver FJ, Helleday T, et al. Contextual synthetic lethality of cancer cell kill based on the tumor microenvironment. Cancer Res. 2010;70(20):8045–8054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Metcalf KJ, Alazzeh A, Werb Z, Weaver VM. Leveraging microenvironmental synthetic lethalities to treat cancer. J Clin Invest. 2021;131(6):e143765. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from BME Frontiers are provided here courtesy of AAAS Science Partner Journal Program

RESOURCES