Engineered ECM models: Opportunities to advance understanding of tumor heterogeneity

https://doi.org/10.1016/j.ceb.2021.04.001Get rights and content

Abstract

Intratumoral heterogeneity is a negative prognostic factor for cancer and commonly attributed to microenvironment-driven genetic mutations and/or the emergence of cancer stem-like cells. How aberrant extracellular matrix (ECM) remodeling regulates the phenotypic diversity of tumor cells, however, remains poorly understood due in part to a lack of model systems that allow isolating the physicochemical heterogeneity of malignancy-associated ECM for mechanistic studies. Here, we review the compositional, microarchitectural, and mechanical hallmarks of cancer-associated ECM and highlight biomaterials and engineering approaches to recapitulate these properties for in vitro and in vivo studies. Subsequently, we describe how such engineered platforms may be explored to define the spatiotemporal dynamics through which cancer-associated ECM remodeling regulates intratumoral heterogeneity and the cancer stem-like cell phenotype. Finally, we highlight future opportunities and technological advances to further elucidate the relationship between tumor-associated ECM dynamics and intratumoral heterogeneity.

Introduction

Intratumoral heterogeneity is a hallmark of cancer and is characterized by the presence of different cancer cell subpopulations that severely limit patient outcomes because of their varied proliferative, invasive, and therapy resistance capabilities [1,2]. Historically, tumor cell heterogeneity has been attributed to oncogenic mutations that increase cell fitness in response to environmental pressures or chemotherapy [3]. However, phenotypic differences caused by epigenetic reprogramming as well as transient changes in gene expression, phosphoproteomics, and metabolic signaling are equally important [2,4,5]. Moreover, the self-renewal and therapy resistance of cancer stem-like cells (CSCs) contribute to clonal diversity within tumors [6]. Indeed, an increase in CSCs because of transformation of tumor cells or environmental selection pressures promotes tumor development, metastasis, and treatment response [6]. Which role the tumor microenvironment (TME) plays in the emergence of CSCs and which effect this has on tumor heterogeneity are not well understood.

Within the TME, cancer cell phenotypes are regulated through crosstalk with tissue-resident stromal cells including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells, and infiltrating immune cells [7]. Much emphasis has been placed on how secretory functions of these cells control tumor heterogeneity and progression. Yet, their impact on the physical properties of the TME may be similarly critical [8]. In particular, CAFs are widely studied for their role in changing the quantity, biochemical composition, and mechanical properties of extracellular matrix (ECM) in tumors [8], and these alterations regulate the genotype and phenotype of tumor cells as well as CSC quantity and functions [9]. Nevertheless, CAF-dependent ECM changes are not homogeneous, but are subject to spatial and temporal variations (Figure 1). How ECM heterogeneity is functionally linked to tumor heterogeneity remains unclear due in part to the lack of relevant model systems.

Both in vivo and in vitro studies have advanced our understanding of how tumor cell interactions with the ECM affect tumor progression and therapy response. However, the high cost and species-dependent differences between humans and mouse models, as well as shortcomings associated with 2-D cell culture, make it challenging to isolate mechanistic links between ECM remodeling and tumor cell state. Engineered model systems can recapitulate and isolate TME-associated ECM changes to probe their effect on tumor heterogeneity as a function of CSC enrichment. Indeed, simply switching tumor cell culture from conventional 2-D to 3-D culture impacts several hallmarks of malignancy, including cellular metabolism [10], invasion [11], and therapy resistance [12]. Furthermore, 3D culturing of cancer cells enriches for CSCs in part through activation of the epithelial-to-mesenchymal transition and altering cytokine secretion [13]. Here, we will summarize current knowledge of ECM changes in the TME, highlight model systems to mimic these changes for mechanistic studies, and outline strategies to further improve the impact engineered ECM models have on our understanding of tumor heterogeneity and the role of CSCs in this process (Figure 1).

Section snippets

Compositional changes

Most prior research studying the role of ECM remodeling in cancer focused on the composition of the ECM and ECM-associated proteins (collectively referred to as the matrisome [14]). Changes in the matrisome relative to healthy tissue are characteristic of aggressive cancers including breast [15] and pancreatic cancer [16]. For example, fibronectin is often increased during tumorigenesis, regulates all stages of the metastatic cascade through integrin-dependent signaling, and impacts CSC marker

Model systems of the extracellular matrix

Decellularized scaffolds generated from tissue, patient samples, or deposited by cells in culture (cell-derived matrices) mimic the native biochemical and physical properties of the ECM (Figure 3a) [23,48, 49, 50]. In particular, CAF-derived cell-derived matrices are often used to recapitulate tumor-associated ECM and promote the malignant potential of both tumor and stromal cells by activating mechanosignaling [49,50]. Despite their obvious benefits, the complexity of decellularized scaffolds

Biomaterial systems to elucidate the interplay between tumor and ECM heterogeneity

Biomaterial models have expanded our understanding of how biophysical alterations of the ECM influence tumor heterogeneity and stemness. For example, studies with PAA and fibrillar collagen gels suggest that ECM stiffness and microarchitecture synergize to increase CSC numbers and tumor metastatic burden [60]. Tumor cells cultured on stiff versus soft PAA gels increase stem cell marker expression and invasiveness, and hypoxia, an independent inducer of tumor cell stemness, further elevates

Conclusion and future perspectives

Studies with engineered ECM models suggest that CSCs interpret biophysical changes in the ECM differently than their differentiated counterparts. These differences may contribute to the phenotypic and genotypic heterogeneity of tumors by inducing the transformation (e.g. through altered mechanotransduction) and selection (e.g. by affecting DNA-damage mechanisms) of tumor cells with stem-like properties. Although these examples highlight how ECM changes alter cell behavior, ECM and cellular

Conflict of interest statement

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

The work described was supported by the Center on the Physics of Cancer Metabolism (1U54CA210184-01) from the National Cancer Institute, a National Science Foundation (NSF) Graduate Research Fellowship (DGE-1650441) to A.A.S, and the Cornell NanoScale Science & Technology Facility (CNF), which is supported by the NSF (NNCI-2025233).

References (79)

  • K.M. Kingsmore et al.

    Interstitial flow differentially increases patient-derived glioblastoma stem cell invasion via CXCR4, CXCL12, and CD44-mediated mechanisms

    Integr Biol

    (2016)
  • B.L. Bangasser et al.

    Shifting the optimal stiffness for cell migration

    Nat Commun

    (2017)
  • D.L. Matera et al.

    Fiber density modulates cell spreading in 3D interstitial matrix mimetics

    ACS Biomater Sci Eng

    (2019)
  • M. Luo et al.

    Distinct FAK activities determine progenitor and mammary stem cell characteristics

    Canc Res

    (2013)
  • M. Luo et al.

    Targeting breast cancer stem cell state equilibrium through modulation of redox signaling

    Cell Metabol

    (2018)
  • M. Gerlinger et al.

    Intratumor heterogeneity and branched evolution revealed by multiregion sequencing

    N Engl J Med

    (2012)
  • A.K. Casasent et al.

    Multiclonal invasion in breast tumors identified by topographic single cell sequencing

    Cell

    (2018)
  • S.M. Shaffer et al.

    Memory sequencing reveals heritable single-cell gene expression programs associated with distinct cellular behaviors

    Cell

    (2020)
  • S. Vasaikar et al.

    Proteogenomic analysis of human colon cancer reveals new therapeutic opportunities

    Cell

    (2019)
  • T. Shibue et al.

    EMT, CSCs, and drug resistance: the mechanistic link and clinical implications

    Nat Rev Clin Oncol

    (2017)
  • F.R. Balkwill et al.

    The tumor microenvironment at a glance

    J Cell Sci

    (2012)
  • E. Sahai et al.

    A framework for advancing our understanding of cancer-associated fibroblasts

    Nat Rev Canc

    (2020)
  • A.S. Cazet et al.

    Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer

    Nat Commun

    (2018)
  • P. DelNero et al.

    3D culture broadly regulates tumor cell hypoxia response and angiogenesis via pro-inflammatory pathways

    Biomaterials

    (2015)
  • A.D. Schwartz et al.

    A biomaterial screening approach reveals microenvironmental mechanisms of drug resistance

    Integr Biol

    (2017)
  • L. Chen et al.

    The enhancement of cancer stem cell properties of MCF-7 cells in 3D collagen scaffolds for modeling of cancer and anti-cancer drugs

    Biomaterials

    (2012)
  • D.E. Desa et al.

    Intratumoral heterogeneity of second-harmonic generation scattering from tumor collagen and its effects on metastatic risk prediction

    BMC Canc

    (2020)
  • C.R. Drifka et al.

    Periductal stromal collagen topology of pancreatic ductal adenocarcinoma differs from that of normal and chronic pancreatitis

    Mod Pathol

    (2015)
  • L.E. Barney et al.

    Tumor cell-organized fibronectin maintenance of a dormant breast cancer population

    Sci Adv

    (2020)
  • S. Jordahl et al.

    Engineered fibrillar fibronectin networks as three-dimensional tissue scaffolds

    Adv Mater

    (2019)
  • K.E. Kubow et al.

    Mechanical forces regulate the interactions of fibronectin and collagen I in extracellular matrix

    Nat Commun

    (2015)
  • N.I. Nissen et al.

    Collagens and Cancer associated fibroblasts in the reactive stroma and its relation to Cancer biology

    J Exp Clin Canc Res

    (2019)
  • A.L. Wishart et al.

    Decellularized extracellular matrix scaffolds identify full-length collagen VI as a driver of breast cancer cell invasion in obesity and metastasis

    Sci Adv

    (2020)
  • B.R. Seo et al.

    Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis

    Sci Transl Med

    (2015)
  • P. Auvinen et al.

    Hyaluronan synthases (HAS1–3) in stromal and malignant cells correlate with breast cancer grade and predict patient survival

    Breast Canc Res Treat

    (2014)
  • M.A. Jacobetz et al.

    Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer

    Gut

    (2013)
  • E. Hammond et al.

    The role of heparanase and sulfatases in the modification of heparan sulfate proteoglycans within the tumor microenvironment and opportunities for novel cancer therapeutics

    Front Oncol

    (2014)
  • S.P. Evanko et al.

    Hyaluronan controls the deposition of fibronectin and collagen and modulates TGF-β1 induction of lung myofibroblasts

    Matrix Biol

    (2015)
  • N. Lohmann et al.

    Glycosaminoglycan-based hydrogels capture inflammatory chemokines and rescue defective wound healing in mice

    Sci Transl Med

    (2017)
  • Cited by (0)

    View full text